1
|
Nepal S, Shi N, Hoyd R, Spakowicz DJ, Orwoll E, Shikany JM, Napoli N, Tabung FK. Role of insulinemic and inflammatory dietary patterns on gut microbial composition and circulating biomarkers of metabolic health among older American men. Gut Microbes 2025; 17:2497400. [PMID: 40296253 PMCID: PMC12045561 DOI: 10.1080/19490976.2025.2497400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/25/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Chronic low-grade inflammation and hyperinsulinemia are linked with metabolic dysfunction and dysbiosis. This study investigated the role of dietary inflammatory and insulinemic potential on gut microbiome and circulating health biomarkers in older men. Data from the Osteoporotic Fractures in Men (MrOS) study were analyzed. Reversed Empirical Dietary Inflammatory Pattern (rEDIP), Empirical Dietary Index for Hyperinsulinemia (rEDIH), and Healthy Eating Index (HEI)-2020 scores were computed from food frequency questionnaire data. Stool samples were profiled using 16S rRNA sequencing. Elastic net regression identified diet-associated microbial profiles and multivariable-adjusted linear regression assessed diet-biomarker associations. Higher rEDIP, rEDIH, and HEI-2020 scores were positively associated with gut microbiota alpha diversity. Specific genera, including Intestinibacter and Lachnospira, associated positively, while Dielma, Peptococcus, Feacalitalea, and Negativibaccilus associated inversely with healthier dietary patterns. When evaluating changes in dietary patterns between baseline and visit 4 ( ~ 14 years), these genera tended to define rEDIP, rEDIH more than HEI-2020. In addition, higher dietary quality was linked to better biomarker profiles, including lower creatinine, sodium, triglycerides, and insulin resistance. Beneficial effects of higher dietary quality on health may be mediated by the ability of diet to regulate gut microbial composition and metabolic biomarker profiles.
Collapse
Affiliation(s)
- Sushma Nepal
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Ni Shi
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Rebecca Hoyd
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Daniel J. Spakowicz
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Eric Orwoll
- Department of Medicine, Oregon Health & Sciences University, Portland, OR, USA
| | - James M. Shikany
- Division of General Internal Medicine and Population Science, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicola Napoli
- Unit of Endocrinology and Diabetes, Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy
- Division of Bone and Mineral Diseases, Washington University, St Louis, MO, USA
| | - Fred K. Tabung
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Division of Epidemiology, College of Public Health, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
2
|
Nagayama M, Gogokhia L, Longman RS. Precision microbiota therapy for IBD: premise and promise. Gut Microbes 2025; 17:2489067. [PMID: 40190259 PMCID: PMC11980506 DOI: 10.1080/19490976.2025.2489067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/19/2024] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
Inflammatory Bowel Disease (IBD) is a spectrum of chronic inflammatory diseases of the intestine that includes subtypes of ulcerative colitis (UC) and Crohn's Disease (CD) and currently has no cure. While IBD results from a complex interplay between genetic, environmental, and immunological factors, sequencing advances over the last 10-15 years revealed signature changes in gut microbiota that contribute to the pathogenesis of IBD. These findings highlight IBD as a disease target for microbiome-based therapies, with the potential to treat the underlying microbial pathogenesis and provide adjuvant therapy to the emerging spectrum of advanced therapies for IBD. Building on the success of fecal microbiota transplantation (FMT) for Clostridioides difficile infection, therapies targeting gut microbiota have emerged as promising approaches for treating IBD; however, unique aspects of IBD pathogenesis highlight the need for more precision in the approach to microbiome therapeutics that leverage aspects of recipient and donor selection, diet and xenobiotics, and strain-specific interactions to enhance the efficacy and safety of IBD therapy. This review focuses on both pre-clinical and clinical studies that support the premise for microbial therapeutics for IBD and aims to provide a framework for the development of precision microbiome therapeutics to optimize clinical outcomes for patients with IBD.
Collapse
Affiliation(s)
- Manabu Nagayama
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
- Jill Roberts Center for Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lasha Gogokhia
- Jill Roberts Center for Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Randy S. Longman
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
- Jill Roberts Center for Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
3
|
Zhao S, Lin H, Li W, Xu X, Wu Q, Wang Z, Shi J, Chen Y, Ye L, Xi L, Chen L, Yuan M, Su J, Gao A, Jin J, Ying X, Wang X, Ye Y, Sun Y, Zhang Y, Deng X, Shen B, Gu W, Ning G, Wang W, Hong J, Wang J, Liu R. Post sleeve gastrectomy-enriched gut commensal Clostridia promotes secondary bile acid increase and weight loss. Gut Microbes 2025; 17:2462261. [PMID: 39915243 PMCID: PMC11810084 DOI: 10.1080/19490976.2025.2462261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 01/07/2025] [Accepted: 01/29/2025] [Indexed: 02/12/2025] Open
Abstract
The gut microbiome is altered after bariatric surgery and is associated with weight loss. However, the commensal bacteria involved and the underlying mechanism remain to be determined. We performed shotgun metagenomic sequencing in obese subjects before and longitudinally after sleeve gastrectomy (SG), and found a significant enrichment in microbial species in Clostridia and bile acid metabolizing genes after SG treatment. Bile acid profiling further revealed decreased primary bile acids (PBAs) and increased conjugated secondary bile acids (C-SBAs) after SG. Specifically, glycodeoxycholic acid (GDCA) and taurodeoxycholic acid (TDCA) were increased at different follow-ups after SG, and were associated with the increased abundance of Clostridia and body weight reduction. Fecal microbiome transplantation with post-SG feces increased SBA levels, and alleviated body weight gain in the recipient mice. Furthermore, both Clostridia-enriched spore-forming bacteria and GDCA supplementation increased the expression of genes responsible for lipolysis and fatty acid oxidation in adipose tissue and reduced adiposity via Takeda G-protein-coupled receptor 5 (TGR5) signaling. Our findings reveal post-SG gut microbiome and C-SBAs as contributory to SG-induced weight loss, in part via TGR5 signaling, and suggest SBA-producing gut microbes as a potential therapeutic target for obesity intervention.
Collapse
Affiliation(s)
- Shaoqian Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huibin Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Qihan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Juan Shi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufei Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingxia Ye
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liuqing Xi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijia Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingyang Yuan
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junlei Su
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aibo Gao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiabin Jin
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiayang Ying
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolin Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yaorui Ye
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yingkai Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifei Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaxing Deng
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baiyong Shen
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Chung J, Pierce J, Franklin C, Olson RM, Morrison AR, Amos-Landgraf J. Translating animal models of SARS-CoV-2 infection to vascular, neurological and gastrointestinal manifestations of COVID-19. Dis Model Mech 2025; 18:dmm052086. [PMID: 40195851 PMCID: PMC12010913 DOI: 10.1242/dmm.052086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
Since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) initiated a global pandemic resulting in an estimated 775 million infections with over 7 million deaths, it has become evident that COVID-19 is not solely a pulmonary disease. Emerging evidence has shown that, in a subset of patients, certain symptoms - including chest pain, stroke, anosmia, dysgeusia, diarrhea and abdominal pain - all indicate a role of vascular, neurological and gastrointestinal (GI) pathology in the disease process. Many of these disease processes persist long after the acute disease has been resolved, resulting in 'long COVID' or post-acute sequelae of COVID-19 (PASC). The molecular mechanisms underlying the acute and systemic conditions associated with COVID-19 remain incompletely defined. Appropriate animal models provide a method of understanding underlying disease mechanisms at the system level through the study of disease progression, tissue pathology, immune system response to the pathogen and behavioral responses. However, very few studies have addressed PASC and whether existing models hold promise for studying this challenging problem. Here, we review the current literature on cardiovascular, neurological and GI pathobiology caused by COVID-19 in patients, along with established animal models of the acute disease manifestations and their prospects for use in PASC studies. Our aim is to provide guidance for the selection of appropriate models in order to recapitulate certain aspects of the disease to enhance the translatability of mechanistic studies.
Collapse
Affiliation(s)
- James Chung
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Julia Pierce
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Department of Research, Ocean State Research Institute, Inc., Providence, RI 02908-4734, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02908, USA
| | - Craig Franklin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Rachel M. Olson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, MO 65211, USA
| | - Alan R. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Department of Research, Ocean State Research Institute, Inc., Providence, RI 02908-4734, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02908, USA
| | - James Amos-Landgraf
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
5
|
Kensiski A, Gavzy SJ, Wu L, Mas V, Ma B, Bromberg JS. Immunosuppressant imprecision: multidirectional effects on metabolism and microbiome. Clin Microbiol Rev 2025; 38:e0017824. [PMID: 40042298 PMCID: PMC12160495 DOI: 10.1128/cmr.00178-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2025] Open
Abstract
SUMMARYTransplant recipients require lifelong, multimodal immunosuppression to prevent rejection by dampening alloreactive immunity. These treatments have long been known to lack antigen specificity. Despite empirically selected long-term immunosuppression regimens, most allografts succumb to alloimmune responses that result in chronic inflammation and scarring. Additionally, immunosuppressive medications themselves contribute to unintended intestinal dysbiosis and metabolic disorders. This review focuses on the effect of immunosuppressant treatments on alloimmunity, gut microbiome, and metabolism, with a particular emphasis on the effects on metabolic disorders. We also outline the shared and unique microbial and metabolic signatures produced by each immunosuppressant class, underlining their distinct impacts on immunity and metabolic homeostasis. These observations underscore the need for a holistic understanding of these drugs' on- and off-target effects to refine therapeutic strategies, enhance immunosuppression efficacy, and ultimately enhance graft and patient survival. By characterizing these complex interactions, strategies informed by the gut microbiome and host metabolism may offer a promising adjunctive approach to optimizing immunosuppressive regimens and promoting sustained graft acceptance.
Collapse
Affiliation(s)
- Allison Kensiski
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Samuel J. Gavzy
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Long Wu
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Valeria Mas
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Bing Ma
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jonathan S. Bromberg
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Yokomizo S, Kaneko N, Chen H, Yan L, Tsuji S, Akagawa S, Sameshima J, Sueyoshi T, Nagano H, Miyahara Y, Kamikaseda Y, Kido H, Hayashi Y, Yamauchi M, Kiyoshima T, Goto Y, Ohyama Y, Kaneko K, Moriyama M, Kawano S. Dysbiosis of the gut microbiome may contribute to the pathogenesis of oral lichen planus through Treg dysregulation. Mucosal Immunol 2025:S1933-0219(25)00055-8. [PMID: 40499683 DOI: 10.1016/j.mucimm.2025.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 05/26/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025]
Abstract
Oral lichen planus (OLP) is a chronic inflammatory disorder with autoimmune features and malignant transformation risk, lacking a definitive treatment, with CD4+ T cells being pivotal in its pathogenesis. Dysbiosis, an imbalance in the microbiome, is linked to various autoimmune and inflammatory diseases, where CD4+ T cells play a significant role. Given these insights, the development of OLP might be influenced by dysbiosis. This study investigates the association between dysbiosis and CD4+ T cells in OLP. We collected stool and saliva samples from OLP patients, conducting 16S rRNA gene analysis and mass spectrometry, and assessed CD4+ T cell characteristics in lesions through multiplex immunofluorescence and single-cell RNA sequencing. Peripheral blood samples were subjected to flow cytometry and cell culture assays. Results showed extensive gut dysbiosis in OLP patients, notably a reduction in short-chain fatty acid (SCFA)-producing bacteria essential for regulatory T cell (Treg) differentiation. While various CD4+ T cell subsets, including Tregs, were present in tissues, these Tregs as unresponsive to specific antigens, showing reduced immunosuppressive molecule expression. The decline in SCFA-producing bacteria correlated with fewer activated Tregs in tissues and blood. These findings suggest that gut dysbiosis may contribute to OLP by impairing Treg regulation, influencing disease pathogenesis.
Collapse
Affiliation(s)
- Shiho Yokomizo
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Naoki Kaneko
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan.
| | - Hu Chen
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Lijing Yan
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Shoji Tsuji
- Department of Pediatrics, Kansai Medical University, Osaka 573-1010, Japan
| | - Shohei Akagawa
- Department of Pediatrics, Kansai Medical University, Osaka 573-1010, Japan
| | - Junsei Sameshima
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Tomoki Sueyoshi
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Haruki Nagano
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Yuka Miyahara
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Yasuhisa Kamikaseda
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Hajime Kido
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshikazu Hayashi
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka 814-0193, Japan; Oral Medicine Research Center, Fukuoka Dental College, Fukuoka 814-0193, Japan
| | - Masaki Yamauchi
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Tamotsu Kiyoshima
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Yuichi Goto
- Department of Maxillofacial Diagnostic and Surgical Science, Field of Oral and Maxillofacial Rehabilitation, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Yukiko Ohyama
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, Osaka 573-1010, Japan
| | - Masafumi Moriyama
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan.
| | - Shintaro Kawano
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
7
|
Lee J, McClure S, Weichselbaum RR, Mimee M. Designing live bacterial therapeutics for cancer. Adv Drug Deliv Rev 2025; 221:115579. [PMID: 40228606 PMCID: PMC12067981 DOI: 10.1016/j.addr.2025.115579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Humans are home to a diverse community of bacteria, many of which form symbiotic relationships with their host. Notably, tumors can also harbor their own unique bacterial populations that can influence tumor growth and progression. These bacteria, which selectively colonize hypoxic and acidic tumor microenvironments, present a novel therapeutic strategy to combat cancer. Advancements in synthetic biology enable us to safely and efficiently program therapeutic drug production in bacteria, further enhancing their potential. This review provides a comprehensive guide to utilizing bacteria for cancer treatment. We discuss key considerations for selecting bacterial strains, emphasizing their colonization efficiency, the delicate balance between safety and anti-tumor efficacy, and the availability of tools for genetic engineering. We also delve into strategies for precise spatiotemporal control of drug delivery to minimize adverse effects and maximize therapeutic impact, exploring recent examples of engineered bacteria designed to combat tumors. Finally, we address the underlying challenges and future prospects of bacterial cancer therapy. This review underscores the versatility of bacterial therapies and outlines strategies to fully harness their potential in the fight against cancer.
Collapse
Affiliation(s)
- Jaehyun Lee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Sandra McClure
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago 60637, USA; The Ludwig Center for Metastasis Research, University of Chicago, Chicago 60637, USA
| | - Mark Mimee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
8
|
Xu Y, Xie R, Weng Y, Fang Y, Tao S, Zhang H, Chen H, Han A, Jiang Q, Liang W. Role and mechanism of gut microbiota-host interactions in the pathogenesis of Crohn's disease. Int J Colorectal Dis 2025; 40:130. [PMID: 40437310 PMCID: PMC12119691 DOI: 10.1007/s00384-025-04917-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2025] [Indexed: 06/01/2025]
Abstract
BACKGROUND Crohn's disease (CD) is a chronic, nonspecific inflammatory bowel disease with a poor prognosis. Despite its increasing incidence, curing CD remains challenging due to its complex etiology and unclear pathogenesis. METHODS A comprehensive PubMed and Web of Science search was conducted using the keywords Crohn's disease, gut microbiota, dysbiosis, pathogenesis and treatment, focusing on studies published between 2014 and 2024. RESULTS Recent studies have demonstrated a close relationship between gut microbiota dysbiosis and the development of CD. Although many dysbioses associated with CD have not yet been proven to be causal or consequential, it has been observed that the gut microbiota in CD patients exhibits reduced diversity, a decrease in beneficial bacteria, and an increase in pathogenic bacteria. These changes may lead to decreased intestinal barrier function, abnormal immune responses, and enhanced inflammatory reactions, which are related to the disease's activity, phenotype, drug treatment efficacy, and postoperative therapeutic outcomes. Therefore, further exploration of the microbiota-host interactions and the pathogenesis of CD, the identification of biomarkers, and the development of targeted strategies for modulating the gut microbiota could offer new avenues for the prevention and treatment of CD. CONCLUSIONS This review highlights the pivotal role of gut microbiota dysbiosis in driving CD pathogenesis and its progression, while underscoring its potential as a therapeutic target through dietary modulation, microbial interventions, and integrative strategies to improve clinical management and prognostic outcomes.
Collapse
Affiliation(s)
- Yao Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| | - Runxiang Xie
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yuqing Weng
- Health Science Center, Ningbo University, Ningbo, China
| | - Yewei Fang
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Shuan Tao
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - He Zhang
- Laboratory Medical School, Bengbu Medical University, Bengbu, China
| | - Huimin Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Axiang Han
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Qi Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| | - Wei Liang
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| |
Collapse
|
9
|
Soumeh EA, Nielsen TS, Hedemann MS, Curtasu MV. Integrated faecal microbiota and blood metabolic changes following different dietary zinc oxide levels in weaned piglets. Sci Rep 2025; 15:18346. [PMID: 40419596 DOI: 10.1038/s41598-025-03103-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Abstract
This study investigated faecal microbial composition and blood metabolome profile of piglets fed different levels of supplementary zinc oxide (ZnO) after weaning. A dose-response study was conducted with four experimental diets containing 153 (D153), 1022 (D1022), 1601 (D1601), and 2407 (D2407) ppm zinc (Zn) in the feed. At the end of the trial, blood and faeces samples were obtained for analyses. Multivariate analysis of the blood metabolomics dataset and Principal Coordinate Analysis (PCoA) of faecal microbiota data showed that pigs receiving D2407 had a different metabolic and microbial profile to the other groups, whereas no differences were observed in pigs fed with D153, D1022, and D1601. The highest dietary Zn inclusion was associated with significant increase in the abundance of Clostridium sensu stricto, Terrisporobacter, Dorea, and Prevotellaceae_NK3B31_group and a decrease in relative abundances of Methanobrevibacter, Treponema, Megasphaera, and UCG 002 genera. Pearson's correlation analysis showed positive correlations between the abundance of Christensenellaceae R7-group with amino acids metabolism and production of microbial metabolites. The results suggest that only 2407 ppm Zn altered gut microbiota and modulated blood metabolic profile, which may impact the health status of piglets through specific microbial metabolites.
Collapse
Affiliation(s)
- Elham Assadi Soumeh
- School of Agriculture and Food Sustainability, Faculty of Science, University of Queensland, Gatton Campus, QLD, 4343, Australia.
| | - Tina Skau Nielsen
- Department of Animal and Veterinary Sciences, Aarhus University AU-Viborg, Blichers Allé 20, Tjele, DK-8830, Denmark
| | - Mette Skou Hedemann
- Department of Animal and Veterinary Sciences, Aarhus University AU-Viborg, Blichers Allé 20, Tjele, DK-8830, Denmark
| | - Mihai Victor Curtasu
- Department of Animal and Veterinary Sciences, Aarhus University AU-Viborg, Blichers Allé 20, Tjele, DK-8830, Denmark
| |
Collapse
|
10
|
Chen Y, Wang Y, Shaoyong W, He Y, Liu Y, Wei S, Gan Y, Sun L, Wang Y, Zong X, Xiang Y, Wang Y, Jin M. High-fertility sows reshape gut microbiota: the rise of serotonin-related bacteria and its impact on sustaining reproductive performance. J Anim Sci Biotechnol 2025; 16:73. [PMID: 40400039 PMCID: PMC12096716 DOI: 10.1186/s40104-025-01191-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/05/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Compelling evidence has established a strong link between the gut microbiota and host reproductive health. However, the specific regulatory roles of individual bacterial species on reproductive performance are not well-understood. In the present study, Jinhua sows with varying reproductive performances under the same diet and management conditions were selected to explore potential mechanisms on the intricate relationship between the gut microbiome and host reproductive performance using 16S rRNA sequencing, metagenomics and serum metabolomics. RESULTS Our findings revealed that the KEGG pathways for base excision repair and DNA replication were enriched, along with gene-level enhancements in spore formation, in sows with higher reproductive performance, indicating that the gut microbiome experiences stress. Further analysis showed a positive correlation between these changes and litter size, indicating that the host acts as a stressor, reshaping the microbiome. This adaptation allows the intestinal microbes in sows with high reproductive performance to enrich specific serotonin-related bacteria, such as Oxalobacter formigenes, Ruminococcus sp. CAG 382, Clostridium leptum, and Clostridium botulinum. Subsequently, the enriched microbiota may promote host serotonin production, which is positively correlated with reproductive performance in our study, known to regulate follicle survival and oocyte maturation. CONCLUSION Our study provides a theoretical basis for understanding the interactions between gut microbes and the host. It highlights new insights into reassembling gut microbiota in sows with higher litter sizes and the role of serotonin-related microbiota and serotonin in fertility.
Collapse
Affiliation(s)
- Yanli Chen
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, China
| | - Yan Wang
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, China
| | - Weike Shaoyong
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, China
| | - Yanmin He
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, China
| | - Yalin Liu
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, China
| | - Siyu Wei
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, China
| | - Yujie Gan
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, China
| | - Lu Sun
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, China
| | - Youming Wang
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, China
| | - Xin Zong
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, China
| | - Yun Xiang
- Institute of Animal Husbandry and Veterinary Medicine, Jinhua Academy of Agricultural Science Research, Jinhua, 321017, China
| | - Yizhen Wang
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, China
| | - Mingliang Jin
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, 310058, China.
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China.
- Zhejiang Key Laboratory of Nutrition and Breeding for High-Quality Animal Products, Hangzhou, 310058, China.
| |
Collapse
|
11
|
Liu Y, Sawada K, Adachi T, Kino Y, Yin T, Yamamoto N, Yamada T. Comparative genome analysis of the immunomodulatory ability of Lactiplantibacillus plantarum and Lactiplantibacillus pentosus from Japanese pickles. mSystems 2025; 10:e0157524. [PMID: 40298372 PMCID: PMC12090711 DOI: 10.1128/msystems.01575-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Lactic acid bacteria (LAB) are pivotal in food preservation and exhibit immunomodulatory effects on interleukin-10 (IL-10) and interleukin-12 (IL-12) production. Lactiplantibacillus plantarum (L. plantarum) and Lactiplantibacillus pentosus (L. pentosus) from fermented food are known for their effect; however, a comprehensive comparative genome analysis is needed to identify the linked genes. Here, we investigated the immunomodulatory capability at the genome level of L. plantarum and L. pentosus strains isolated from Japanese pickles at the genome level, and we further identified their immunomodulation-associated genes using the potential-gene (PG) index derived from the Calinski-Harabasz (CH) index. The results revealed an immunostimulatory clade with strain-specific IL-10 and IL-12 induction and identified key genes via the PG index. Both genes across two species were shown to encode the enzyme TagF2, which is crucial for synthesizing poly-glycerol-3-phosphate type wall teichoic acid (poly-GroP WTA), indicating that TagF2 plays a potential role as an effective microbe-associated-molecular-pattern. In vivo analyses confirmed the IL-10-inducing ability of one strain, reinforcing the IL-10-stimulating capacity of its poly-GroP WTA. Subpotential genes in L. plantarum TagF2-possessing strains were linked to host‒cell interactions, suggesting that such strains play potential probiotic roles. Collectively, the PG index effectively identified immunomodulation-related genes, thus paving the way for the use of the PG index to detect potential health benefit-associated genes in other LAB species. IMPORTANCE Lactic acid bacteria are pivotal in food preservation and exhibit immunomodulatory effects on interleukin-10 (IL-10) and interleukin-12 (IL-12) production. Lactiplantibacillus plantarum and Lactiplantibacillus pentosus from fermented food are known for such effect, yet comprehensive comparative genome analysis is needed to elucidate the linked genes of the two species. The significance of our research is in observing the immunostimulatory clade with strain-specific cytokine induction and identifying key immunostimulation-related genes encoding enzymes that are crucial for synthesizing a potentially effective microbe-associated-molecular-pattern using the potential-gene index across two species. The further in vivo validation reinforced the interleukin-10-stimulating capacity of the identified pattern, and the detected sub-potential genes in Lactiplantibacillus plantarum key-gene possessing strains implied the utility of potential-gene index in detecting potential health-benefit-associated genes in other lactic acid bacteria species.
Collapse
Affiliation(s)
- Yiting Liu
- School of Life Science and Technology, Institute of Science Tokyo, Tokyo, Japan
| | | | - Takahiko Adachi
- Department of Precision Health, Medical Research Institute, The Institute of Medical Engineering, Institute of New Industry Incubation, Institute of Science Tokyo, Tokyo, Japan
| | - Yuta Kino
- School of Life Science and Technology, Institute of Science Tokyo, Tokyo, Japan
| | - Tingyu Yin
- School of Life Science and Technology, Institute of Science Tokyo, Tokyo, Japan
| | - Naoyuki Yamamoto
- School of Life Science and Technology, Institute of Science Tokyo, Tokyo, Japan
- Laboratory for Intestinal Microbiota, Juntendo University, Bunkyo City, Tokyo, Japan
| | - Takuji Yamada
- School of Life Science and Technology, Institute of Science Tokyo, Tokyo, Japan
- Metagen, Inc., Yamagata, Japan
- Metagen Theurapeutics, Inc., Yamagata, Japan
- digzyme, Inc., Tokyo, Japan
| |
Collapse
|
12
|
Kopa-Stojak PN, Kleniewska P, Hoffmann A, Pawliczak R. The potential effect of gut microbiota on the secretion of selected cytokines by human cells in vitro. Sci Rep 2025; 15:17367. [PMID: 40389545 PMCID: PMC12089611 DOI: 10.1038/s41598-025-01581-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
Colonization of the gut early in life plays a significant role in guiding the development of the immune system. The effect of individual intestinal bacterial strains on the asthma development is the subject of numerous scientific studies. The aim of the study was to determine the role and the potential mechanism of action of intestinal microflora on secretion of cytokines and potential predisposition to asthma development. The effect of Parabacteroides distasonis (PD), Bacteroides vulgatus (BV) Clostridium perfringens (CP) and Ruminococcus albus (RA) lysates on the secretion of IL-4, IL-5, IL-8 (CXCL8) and IL-13 in human peripheral blood mononuclear cells, monocytes and HT-29 cells has been analyzed by enzyme-linked immunosorbent assays. RA and PD 400 µg lysates significantly increased secretion of IL-5 by PBMC compared to control (p < 0.05). In addition, BV, CP, PD and RA 100 µg lysates significantly increased IL-8 secretion by PBMC, as well as BV, PD and RA 100 µg lysates by monocytes compared to control (p < 0.05). Moreover, PD 100 µg and 400 µg lysates significantly increased secretion of IL-8 by HT-29 cells compared to BV 100 µg and 400 µg lysates (p < 0.05). CP, BV, PD and RA 100 µg lysates significantly lowered IL-13 secretion by PBMC vs. control (p < 0.001). For a better understanding of the mechanisms of action of gut microflora and their impact on parameters important in asthma, complex studies which compare the asthma and control samples should be carried out in the future.
Collapse
Affiliation(s)
- Paulina Natalia Kopa-Stojak
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Zeligowskiego 7/9, (bldg 2 Rm 177), 90-752, Lodz, Poland
| | - Paulina Kleniewska
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Zeligowskiego 7/9, (bldg 2 Rm 177), 90-752, Lodz, Poland
| | - Arkadiusz Hoffmann
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Zeligowskiego 7/9, (bldg 2 Rm 177), 90-752, Lodz, Poland
| | - Rafał Pawliczak
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Zeligowskiego 7/9, (bldg 2 Rm 177), 90-752, Lodz, Poland.
| |
Collapse
|
13
|
Qiao Y, Zheng H, Cheng R, Guo J, Ji L, Liu Z, Wei L, Bi H, Shan Z. High-fat diet-induced osteoporosis in mice under hypoxic conditions. BMC Musculoskelet Disord 2025; 26:487. [PMID: 40380162 PMCID: PMC12082882 DOI: 10.1186/s12891-025-08725-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
In the context of global aging, osteoporosis has emerged as a significant public health concern, with a relatively high prevalence observed in plateau regions. This study aimed to investigate the effects and underlying mechanisms of high-fat diet (HFD) and hypoxic conditions on bone metabolism in mice. The mice were subjected to different dietary regimens (a HFD versus a normal diet) and placed in a hypoxic environment. This study explored relevant mechanisms through comprehensive assessments, including body and bone morphological indices, pathological examinations, biochemical analyses, evaluation of gut microbiota diversity, and metabolomics approaches. The results indicated that, compared with those in the control group, the body weight, Lee's index, body mass index (BMI), and body fat percentage of the HFD-fed group were significantly greater. Additionally, the femoral microstructure was compromised, bone metabolic markers were disrupted, inflammatory responses were heightened, gut microbiota diversity was altered, and specific intestinal metabolites such as Anserine were downregulated, whereas L-carnosine was upregulated. Spearman correlation analysis and network visualization elucidated the multifactorial influence mechanism of a HFD on bone metabolism under hypoxic conditions. These factors interconnect to form a complex network that drives osteoporosis development. Notably, L-carnosine occupies a central position within this network, serving as a key hub for interactions among various factors. Under the dual stressors of hypoxia and a HFD, this network becomes imbalanced, leading to bone metabolic disorders and osteoporosis. This study provides insights into the multifactorial mechanisms of osteoporosis induced by a HFD and hypoxia in mice, offering a foundation for subsequent research and preventive strategies for osteoporosis in plateau areas.
Collapse
Affiliation(s)
- Yajun Qiao
- Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, Chinese Academy of Science, 23 Xinning Road, Xining, 810001, China
- University of Chinese Academy of Sciences, 19(A) yuquan road, Beijing, 10049, China
| | - Huimin Zheng
- Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, Chinese Academy of Science, 23 Xinning Road, Xining, 810001, China
- Department of Pharmacy, Qinghai Minzu University, Xining, 810007, China
| | - Ruiying Cheng
- Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, Chinese Academy of Science, 23 Xinning Road, Xining, 810001, China
- University of Chinese Academy of Sciences, 19(A) yuquan road, Beijing, 10049, China
| | - Juan Guo
- Department of Orthopedic Surgery, People's Hospital of Qinghai Province, 2 Gonghe Road, Xining, 810007, China
- University of Chinese Academy of Sciences, 19(A) yuquan road, Beijing, 10049, China
- Xining Chengxi District Center for Disease Control and Prevention, Fuxing Lane 8, Xining, 810007, China
| | - Li Ji
- Department of Orthopedic Surgery, People's Hospital of Qinghai Province, 2 Gonghe Road, Xining, 810007, China
| | - Zhibin Liu
- Department of Orthopedic Surgery, People's Hospital of Qinghai Province, 2 Gonghe Road, Xining, 810007, China
| | - Lixin Wei
- Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, Chinese Academy of Science, 23 Xinning Road, Xining, 810001, China
- University of Chinese Academy of Sciences, 19(A) yuquan road, Beijing, 10049, China
| | - Hongtao Bi
- Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, Chinese Academy of Science, 23 Xinning Road, Xining, 810001, China.
- University of Chinese Academy of Sciences, 19(A) yuquan road, Beijing, 10049, China.
| | - Zhongshu Shan
- Department of Orthopedic Surgery, People's Hospital of Qinghai Province, 2 Gonghe Road, Xining, 810007, China.
| |
Collapse
|
14
|
Yadav SK, Chen C, Dhib-Jalbut S, Ito K. The mechanism of disease progression by aging and age-related gut dysbiosis in multiple sclerosis. Neurobiol Dis 2025; 212:106956. [PMID: 40383164 DOI: 10.1016/j.nbd.2025.106956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 05/05/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025] Open
Abstract
Multiple sclerosis (MS) is the most common demyelinating disease caused by a multifaceted interplay of genetic predispositions and environmental factors. Most patients initially experience the relapsing-remitting form of the disease (RRMS), which is characterized by episodes of neurological deficits followed by periods of symptom resolution. However, over time, many individuals with RRMS advance to a progressive form of the disease, known as secondary progressive MS (SPMS), marked by a gradual worsening of symptoms without periods of remission. The mechanisms underlying this transition remain largely unclear, and current disease-modifying therapies (DMTs) are partially effective in treating SPMS. Age is widely acknowledged as a risk factor for the transition from RRMS to SPMS. One factor associated with aging that may influence the progression of MS is gut dysbiosis. This review discusses how aging and age-related gut dysbiosis affect the progression of MS.
Collapse
Affiliation(s)
- Sudhir Kumar Yadav
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, United States of America
| | - Claire Chen
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, United States of America
| | - Suhayl Dhib-Jalbut
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, United States of America
| | - Kouichi Ito
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, United States of America.
| |
Collapse
|
15
|
Teuscher JL, Lupatsii M, Graspeuntner S, Jonassen S, Bringewatt A, Herting E, Stichtenoth G, Bossung V, Rupp J, Härtel C, Demmert M. Persistent reduction of Bifidobacterium longum in the infant gut microbiome in the first year of age following intrapartum penicillin prophylaxis for maternal GBS colonization. Front Immunol 2025; 16:1540979. [PMID: 40443663 PMCID: PMC12119681 DOI: 10.3389/fimmu.2025.1540979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/23/2025] [Indexed: 06/02/2025] Open
Abstract
Introduction Group B Streptococcus is a significant cause of early-onset disease in term newborns, with a global incidence of 0.41/1000 live births. Intrapartum antibiotic prophylaxis (IAP) has reduced EOD incidence by over 80%, but concerns exist about its impact on the neonatal gut microbiome and potential long-term health effects. Methods This single center study examines the effects of IAP on the fecal infant microbiome in the first year of age and on the T cell phenotype in the first days after birth among 22 infants receiving IAP with penicillin due to maternal GBS colonization and 26 infants not exposed to IAP. The fecal microbiome was analyzed at birth, one month and one year of age through 16S rRNA gene sequencing. Additionally, a T cell phenotyping of peripheral blood was performed between the second and fifth day of age. Results At one month, IAP exposed infants had a significantly lower relative abundance of Bifidobacterium longum in fecal samples, an effect which was sustained at one year. In IAP exposed infants we found a proinflammatory T-helper cell profile, characterized by higher IL-17A, RORgt, and TGF-b expression. Discussion This study proposes a sustained impact of IAP on the neonatal microbiome and T cell repertoire.
Collapse
Affiliation(s)
- Jana Lucia Teuscher
- Clinic for Pediatric and Adolescent Medicine, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Mariia Lupatsii
- Department for Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Simon Graspeuntner
- Department for Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein, Lübeck, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
- Medical Clinic III, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Sinje Jonassen
- Clinic for Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Arne Bringewatt
- Clinic for Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Egbert Herting
- Clinic for Pediatric and Adolescent Medicine, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Guido Stichtenoth
- Clinic for Pediatric and Adolescent Medicine, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Verena Bossung
- Clinic for Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Jan Rupp
- Department for Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein, Lübeck, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Christoph Härtel
- Pediatric Clinic and Policlinic, University Hospital Würzburg, Würzburg, Germany
| | - Martin Demmert
- Clinic for Pediatric and Adolescent Medicine, University Hospital Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|
16
|
Andreou E, Papaneophytou C. Boosting Immunity Through Nutrition and Gut Health: A Narrative Review on Managing Allergies and Multimorbidity. Nutrients 2025; 17:1685. [PMID: 40431425 PMCID: PMC12114198 DOI: 10.3390/nu17101685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
The increasing global burden of allergic diseases and multimorbidity underscores the urgent need for innovative strategies to strengthen immune health. This review explores the complex relationships among nutrition, gut microbiota, immune regulation, allergic diseases, and multimorbidity. It highlights how targeted nutritional and microbial interventions may influence disease outcomes. Dietary components and microbial metabolites dynamically modulated immune function, highlighting the critical role of the gut-immune-metabolism axis in disease pathogenesis and management. Personalized nutrition, guided by advances in diagnostics such as component-resolved diagnostics, basophil activation tests, and epigenetic biomarkers, allows for precise dietary interventions tailored to individual allergy phenotypes and multimorbidity profiles. The Mediterranean diet, breastfeeding, and microbiota-targeted therapies have emerged as effective strategies to enhance immune resilience, reduce inflammation, and manage allergic reactions. Technological advancements, including artificial intelligence-driven dietary assessments, wearable devices, and mobile applications, have further revolutionized personalized dietary management, enabling real-time, precise nutritional monitoring and intervention. Despite these advances, challenges in implementing personalized nutrition persist, including variability in dietary patterns, cultural and socioeconomic factors, and accessibility concerns. Future research should focus on long-term interventional and longitudinal studies to validate precision nutrition strategies and enhance clinical applicability. This integrative approach, combining nutrition, microbiome science, technology, and personalized healthcare, holds substantial promises for sustainable disease prevention and enhanced immune resilience across diverse populations.
Collapse
Affiliation(s)
| | - Christos Papaneophytou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus;
| |
Collapse
|
17
|
Yi J, Jung J, Horton D, Hsieh P, Peng Y, Wang SJ, Newberry R, Ericsson AC, Kim KS, Kau AL, Hsieh CS. A hierarchy of intestinal antigens instructs the CD4 + T cell receptor repertoire. Immunity 2025; 58:1217-1235.e4. [PMID: 40318631 DOI: 10.1016/j.immuni.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/12/2024] [Accepted: 04/10/2025] [Indexed: 05/07/2025]
Abstract
Intestinal CD4+ T cells that are specific for self-, diet-, or commensal-derived antigens are critical for host tolerance but must also be tightly regulated to prevent aberrant activation and conditions like inflammatory bowel disease (IBD). However, it is unclear how the antigen source and location dictate the intestinal TCR repertoire. Here, we hierarchically classified self-, diet-, or microbiota-dependent TCRs using TCliβ TCRβ transgenic mice. This demonstrated that microbiota had a greater influence than diet on CD4+ T cell responses throughout the intestine at homeostasis. Complex bi-directional interactions between microbes and diet were also observed. In the context of murine colitis as a model of IBD, we showed that antigen-free diet substantially altered the microbiota and associated T cell responses, which ameliorated intestinal inflammation. Collectively, these findings suggest how deconvoluting the gut immune interactome may facilitate identifying primary microbial and dietary drivers of T cell responses during health and disease.
Collapse
MESH Headings
- Animals
- Mice
- CD4-Positive T-Lymphocytes/immunology
- Mice, Transgenic
- Gastrointestinal Microbiome/immunology
- Mice, Inbred C57BL
- Inflammatory Bowel Diseases/immunology
- Inflammatory Bowel Diseases/microbiology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Antigens/immunology
- Colitis/immunology
- Colitis/microbiology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Intestinal Mucosa/immunology
- Disease Models, Animal
- Intestines/immunology
- Intestines/microbiology
Collapse
Affiliation(s)
- Jaeu Yi
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biological Science, Ajou University, Suwon 16499, Republic of Korea
| | - Jisun Jung
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David Horton
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Patricia Hsieh
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yangqing Peng
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sean J Wang
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rodney Newberry
- Department of Internal Medicine, Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Aaron C Ericsson
- The Mutant Mouse Resource and Research Center, University of Missouri (MU-MMRRC), Columbia, MO 65201, USA
| | - Kwang Soon Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Andrew L Kau
- Division of Allergy and Immunology, Department of Medicine and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chyi-Song Hsieh
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
18
|
Kim SH, White Z, Gainullina A, Kang S, Kim J, Dominguez JR, Choi Y, Cabrera I, Plaster M, Takahama M, Czepielewski RS, Yeom J, Gunzer M, Hay N, David O, Chevrier N, Sano T, Kim KW. IL-10 sensing by lung interstitial macrophages prevents bacterial dysbiosis-driven pulmonary inflammation and maintains immune homeostasis. Immunity 2025; 58:1306-1326.e7. [PMID: 40306274 DOI: 10.1016/j.immuni.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 10/02/2024] [Accepted: 04/03/2025] [Indexed: 05/02/2025]
Abstract
Crosstalk between the immune system and the microbiome is critical for maintaining immune homeostasis. Here, we examined this communication and the impact of immune-suppressive IL-10 signaling on pulmonary homeostasis. We found that IL-10 sensing by interstitial macrophages (IMs) is required to prevent spontaneous lung inflammation. Loss of IL-10 signaling in IMs initiated an inflammatory cascade through the activation of classical monocytes and CD4+ T cell subsets, leading to chronic lung inflammation with age. Analyses of antibiotic-treated and germ-free mice established that lung inflammation in the animals lacking IL-10 signaling was triggered by commensal bacteria. 16S rRNA sequencing revealed Delftia acidovorans and Rhodococcus erythropolis as potential drivers of lung inflammation. Intranasal administration of these bacteria or transplantation of human fecal microbiota elicited lung inflammation in gnotobiotic Il10-deficient mice. These findings highlight that IL-10 sensing by IMs contributes to pulmonary homeostasis by preventing lung inflammation caused by commensal dysbiosis.
Collapse
Affiliation(s)
- Seung Hyeon Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Zachary White
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
| | | | - Soeun Kang
- Department of Biochemistry and Genetics, University of Illinois College of Medicine, Chicago, IL, USA
| | - Jiseon Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Joseph R Dominguez
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Yeonwoo Choi
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Ivan Cabrera
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Madison Plaster
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Michihiro Takahama
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Rafael S Czepielewski
- Immunology Center of Georgia, Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Jinki Yeom
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Nissim Hay
- Department of Biochemistry and Genetics, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Odile David
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA; Department of Pathology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Nicolas Chevrier
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Teruyuki Sano
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| | - Ki-Wook Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
19
|
Uchida A, Imai K, Miki R, Hamaguchi T, Nishiwaki H, Ito M, Ueyama J, Hattori S, Tano S, Fuma K, Matsuo S, Ushida T, Ohno K, Kajiyama H, Kotani T. Butyrate-producing bacteria in pregnancy maintenance: mitigating dysbiosis-induced preterm birth. J Transl Med 2025; 23:533. [PMID: 40355924 PMCID: PMC12067704 DOI: 10.1186/s12967-025-06534-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Preterm birth (PTB) is a major contributor to neonatal morbidity, mortality, and long-term health complications. Despite advances in perinatal care, PTB rates remain high, and its multifactorial etiology is not fully understood. Increasing evidence suggests that maternal gut microbiota plays a critical role in pregnancy maintenance, potentially through modulation of immune responses. However, the underlying causal mechanisms remain unclear. We hypothesized that dysbiosis disrupts immune tolerance and promotes PTB, and that butyrate (short-chain fatty acid produced by specific gut bacteria) may counteract this effect by enhancing regulatory T cell (Treg)-mediated immune regulation. METHODS We established a dysbiosis-induced PTB mouse model using vancomycin treatment combined with subclinical immune activation via anti-CD3ε antibody. Pregnant mice were fed either a standard or butyrate-enriched diet. Outcomes included gestational length, PTB incidence, live pup rates, and Treg cell levels assessed by flow cytometry. Parallelly, 16S rRNA gene sequencing was performed on fecal samples from 32 pregnant women to compare gut microbial composition between spontaneous PTB and term birth groups. Multivariate logistic regression and correlation analyses were conducted to assess associations with gestational outcomes. RESULTS Vancomycin-induced dysbiosis in mice significantly reduced Treg cell populations and increased PTB rates (43.3% in dysbiosis vs. 0% in controls; p < 0.05), while butyrate supplementation reduced PTB incidence (p = 0.03), prolonged gestation (p = 0.01), and restored Treg counts (p < 0.001). In human samples, significant reductions in Lachnospiraceae and Ruminococcaceae, representative butyrate-producing bacteria, were seen in PTB cases. Their combined abundance was independently associated with sPTB risk (p = 0.019) and positively correlated with gestational age (r = 0.59, p < 0.001). CONCLUSIONS Our findings demonstrate that maternal dysbiosis increases PTB risk via impaired immune tolerance, and that butyrate supplementation effectively reverses this effect in vivo. Human data support the translational relevance of butyrate-producing microbiota in pregnancy maintenance. These results highlight butyrate as a promising target for dietary interventions aimed at reducing PTB incidence by restoring immune homeostasis. Trial registration Not applicable.
Collapse
Affiliation(s)
- Azusa Uchida
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kenji Imai
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Rika Miki
- Maternal Fetal Health Laboratory, Research Institute, Nozaki Tokushukai Hospital, Daito, Osaka, Japan
- Laboratory of Bell Research Center‑Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomonari Hamaguchi
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Nishiwaki
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun Ueyama
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satomi Hattori
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Sho Tano
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kazuya Fuma
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Seiko Matsuo
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Takafumi Ushida
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomomi Kotani
- Departments of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
20
|
Sharif-Askari EA, Atoui KM, Mteyrek AK, Fawaz LM. Probiotics and mediterranean diet for breast cancer management and prevention? Cell Stress 2025; 9:1-15. [PMID: 40417456 PMCID: PMC12096334 DOI: 10.15698/cst2025.05.303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2025] [Revised: 01/30/2025] [Accepted: 03/25/2025] [Indexed: 05/27/2025] Open
Abstract
The human gut microbiota, a diverse community of beneficial normal flora microorganisms, significantly influences physiological function and the immune response. Various microbiota strains have shown promise in supporting clinical treatment of chronic diseases, including cancer, by potentially providing antioxidative and anti-tumorigenic effects in both in vivo and in vitro studies. Breast cancer, which ranks amongst the top five cancer types common worldwide and particularly in Mediterranean countries, has been showing high incidence and prevalence. In breast cancer, microbiota composition, hormonal dynamics, and dietary choices are believed to play significant roles. Hence, the Mediterranean diet, known for its microbiota-friendly features, emerges as a potential protective factor against breast cancer development, highlighting the potential for personalized dietary strategies in cancer prevention. This comprehensive review highlights the emerging mechanisms by which probiotics support our immune system during different physiological activities. It also discusses their potential role, along with nutrition intervention, in improving essential clinical treatment outcomes in breast cancer patients and survivors, suggesting potential supportive strategies that go hand in hand with clinical strategies. Unfortunately, very little research addresses the possible clinical implications of probiotics and dietary habits on breast cancer, despite the promising results, calling for further studies and actions.
Collapse
Affiliation(s)
- Ehssan A. Sharif-Askari
- Biomedical Science Department, School of Arts & Sciences, Lebanese International University, Tyre, Lebanon
| | - Khadija M. Atoui
- Biomedical Science Department, School of Arts & Sciences, Lebanese International University, Tyre, Lebanon
| | - Ali K. Mteyrek
- Department of Biological and Chemical Sciences, School of Arts and Sciences, Lebanese International University, Tyre, Lebanon
| | - Lama M. Fawaz
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Quebec, Canada
| |
Collapse
|
21
|
Huang Y, Fang R, Xiong T, Li W, Yu N. The Relationship Between Gut Microbiota and Recurrent Spontaneous Abortion. Microorganisms 2025; 13:1073. [PMID: 40431246 PMCID: PMC12114443 DOI: 10.3390/microorganisms13051073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/20/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
Recently, the gut microbiota has been found to be associated with multiple organs and systems in the human body, playing a key role in the occurrence and development of various diseases, such as the gut-brain axis and the gut-liver axis. However, its interaction with miscarriages remains poorly understood. This article reviews the characteristics of gut microbiota and its metabolites in patients with recurrent spontaneous abortion (RSA), the mechanism of gut microbiota inducing RSA, and potential therapeutic strategies. Therefore, it provides a new perspective on the gut microbiota in the pathogenesis and treatment of recurrent abortion, and the prospect of the future research direction of gut microbiota and recurrent abortion is proposed based on existing studies.
Collapse
Affiliation(s)
- Yiyao Huang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (Y.H.); (R.F.)
| | - Ruijie Fang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (Y.H.); (R.F.)
| | - Ting Xiong
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (T.X.); (W.L.)
| | - Wei Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (T.X.); (W.L.)
| | - Nan Yu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (T.X.); (W.L.)
| |
Collapse
|
22
|
Wang J, Cheng W, Yang R. Nervous system-gut microbiota-immune system axis: future directions for preventing tumor. Front Immunol 2025; 16:1535955. [PMID: 40376000 PMCID: PMC12078214 DOI: 10.3389/fimmu.2025.1535955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/01/2025] [Indexed: 05/18/2025] Open
Abstract
Tumor is one of the leading causes of death worldwide. The occurrence and development of tumors are related to multiple systems and factors such as the immune system, gut microbiota, and nervous system. The immune system plays a critical role in tumor development. Studies have also found that the gut microbiota can directly or indirectly affect tumorigenesis and tumor development. With increasing attention on the tumor microenvironment in recent years, the nervous system has emerged as a novel regulator of tumor development. Some tumor therapies based on the nervous system have also been tested in clinical trials. However, the nervous system can not only directly interact with tumor cells but also indirectly affect tumor development through the gut microbiota. The nervous system-mediated gut microbiota can regulate tumorigenesis, growth, invasion, and metastasis through the immune system. Here, we mainly explore the potential effects of the nervous system-gut microbiota-immune system axis on tumorigenesis and tumor development. The effects of the nervous system-gut microbiota-immune system axis on tumors involve the nervous system regulating immune cells through the gut microbiota, which can prevent tumor development. Meanwhile, the direct effects of the gut microbiota on tumors and the regulation of the immune system by the nervous system, which can affect tumor development, are also reviewed.
Collapse
Affiliation(s)
- Juanjuan Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Wenyue Cheng
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
| |
Collapse
|
23
|
Hou L, Fu Y, Zhao C, Fan L, Hu H, Yin S. Short-term exposure to ciprofloxacin and microplastic leads to intrahepatic cholestasis, while long-term exposure decreases energy metabolism and increases the risk of obesity. ENVIRONMENT INTERNATIONAL 2025; 199:109511. [PMID: 40328087 DOI: 10.1016/j.envint.2025.109511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/02/2025] [Accepted: 04/29/2025] [Indexed: 05/08/2025]
Abstract
Microplastics (MPs) and antibiotics are pervasive pollutants that may pose a risk to human health. Studies have shown that both MPs and antibiotics adversely affect lipid metabolism and increase the risk of obesity. However, it remains unclear whether combined exposure to these pollutants intensify the cumulative detrimental effect on obesity and metabolism. This study demonstrated the impact of exposure to polystyrene MPs (PS, 25 nm) and ciprofloxacin (CIP), both individually and combined, for 30 d and 90 d on the hepatic metabolism of male C57BL/6J mice. The results showed that mice exposed to PS and CIP for either 30 d or 90 d exhibited lipid metabolism disorders such as increased body weight, enlarged adipocytes, triglyceride accumulation in the liver, and higher HDL-C. Differentially expressed hepatic proteins were identified via proteomic analysis. The findings indicated that exposure for 30 d caused abnormal bile acid (BA) secretion in the liver and inhibited the BA secretion pathway, which resulted in intrahepatic cholestasis. Furthermore, exposure for 90 d resolved cholestasis and reduced the overall number of differentially expressed proteins. Intestinal pathology revealed more severe damage after exposure for 30 d, while 90 d exposure decreased the adverse effect. Combined CIP and PS exposure caused damage to the organism. However, the adaptive capacity of the organism during prolonged exposure mitigated the damage caused by both, but did not imply the complete eradication of adverse effects. This study found that 90 d exposure to PS and CIP resulted in weight gain, possibly due to changes in the gut flora and suppressed energy metabolism. These results indicated that simultaneous exposure to CIP and PS exacerbated the adverse impact on the liver, causing short-term intrahepatic cholestasis. Prolonged exposure reduced the energy metabolism in the body, exhibiting varied toxicity outcomes and mechanisms at different exposure durations. This study offers novel insights into the effect of MPs and antibiotic CIP exposure on metabolic abnormalities and provides a scientific basis for assessing these risks. It also emphasizes that the adverse effect resulting from 30 d (short-term) toxic exposure may not persist and that long-term chronic toxicity needs warrants.
Collapse
Affiliation(s)
- Lirui Hou
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Yuhan Fu
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Chong Zhao
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Lihong Fan
- College of Veterinary Medicine, China Agricultural University, Yunamingyuan West Road, Haidian District, Beijing 100193, China
| | - Hongbo Hu
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Shutao Yin
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua East Road, Haidian District, Beijing 100083, China.
| |
Collapse
|
24
|
Husain N, Kumar A, Anbazhagan AN, Gill RK, Dudeja PK. Intestinal luminal anion transporters and their interplay with gut microbiome and inflammation. Am J Physiol Cell Physiol 2025; 328:C1455-C1472. [PMID: 40047092 PMCID: PMC12023768 DOI: 10.1152/ajpcell.00026.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/29/2025] [Accepted: 02/28/2025] [Indexed: 04/16/2025]
Abstract
The intestine, as a critical interface between the external environment and the internal body, plays a central role in nutrient absorption, immune regulation, and maintaining homeostasis. The intestinal epithelium, composed of specialized epithelial cells, harbors apical anion transporters that primarily mediate the transport of chloride and bicarbonate ions, essential for maintaining electrolyte balance, pH homeostasis, and fluid absorption/secretion. In addition, the intestine hosts a diverse population of gut microbiota that plays a pivotal role in various physiological processes including nutrient metabolism, immune regulation, and maintenance of intestinal barrier integrity, all of which are critical for host gut homeostasis and health. The anion transporters and gut microbiome are intricately interconnected, where alterations in one can trigger changes in the other, leading to compromised barrier integrity and increasing susceptibility to pathophysiological states including gut inflammation. This review focuses on the interplay of key apical anion transporters including Down-Regulated in Adenoma (DRA, SLC26A3), Putative Anion Transporter-1 (PAT1, SLC26A6), and Cystic Fibrosis Transmembrane Conductance Regulator [CFTR, ATP-binding cassette subfamily C member 7 (ABCC7)] with the gut microbiome, barrier integrity, and their relationship to gut inflammation.
Collapse
Affiliation(s)
- Nazim Husain
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
| | - Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Arivarasu N. Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
| | - Ravinder K Gill
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Pradeep. K. Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
25
|
Dong Z, Zhang R, Shen L, Ji H, He H, Ji X, Zhao L. Gut Microbiota and Immunoglobulin A Nephropathy: Exploration of Dietary Intervention and Treatment Strategies. Food Sci Nutr 2025; 13:e70218. [PMID: 40321610 PMCID: PMC12045934 DOI: 10.1002/fsn3.70218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
Immunoglobulin A nephropathy (IgAN) is a primary glomerular disease characterized by the deposition of IgA. The pathogenesis of it is related to the dysbiosis of gut microbiota. Dysbiosis of gut microbiota influences mucosal immune response and systemic immune system, leading to glycosylation-deficient IgA1 (Gd-IgA1) increasing, which promotes the development of IgAN. Diet plays an important role in regulating gut microbiota and treating IgAN. In this review, we summarize the interplay between gut microbiota and IgAN, and their underlying mechanisms. We also describe the effects of dietary intake on IgAN, as well as the composition of gut microbiota. The progress on IgAN treatment mainly focuses on inhibiting or regulating the immune system. Moreover, therapeutic strategies related to gut microbiota such as dietary intervention, supplement of probiotics and prebiotics, as well as fecal microbiota transplantation (FMT) have shown the possibility of improving IgAN prognosis. Thus, exploration of the gut-kidney axis, the long-term effects of diet and microbiome is necessary to develop more effective treatment strategies.
Collapse
Affiliation(s)
- Zhaoyang Dong
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiChina
| | - Ran Zhang
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiChina
| | - Liang Shen
- Institute of Food and Drug Research for One Health, School of Food EngineeringLudong UniversityYantaiPeople's Republic of China
| | - Hong‐Fang Ji
- Institute of Food and Drug Research for One Health, School of Food EngineeringLudong UniversityYantaiPeople's Republic of China
| | - Haidong He
- Department of NephrologyMinhang Hospital, Fudan UniversityShanghaiChina
| | - Xiaoguo Ji
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiChina
- Shanghai Frontiers Science Centre of Optogenetic Techniques for Cell Metabolism, School of PharmacyEast China University of Science and TechnologyShanghaiChina
| | - Liming Zhao
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiChina
- Shanghai Collaborative Innovation Centre for Biomanufacturing Technology (SCICBT)ShanghaiChina
| |
Collapse
|
26
|
Yu Z, Wang Y, Guo Y, Zhu R, Fang Y, Yao Q, Fu H, Zhou A, Ma L, Shou Q. Exploring the Therapeutic and Gut Microbiota-Modulating Effects of Qingreliangxuefang on IMQ-Induced Psoriasis. Drug Des Devel Ther 2025; 19:3269-3291. [PMID: 40322026 PMCID: PMC12048299 DOI: 10.2147/dddt.s492044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Purpose To investigate the therapeutic and gut microbiota-modulating effects of Qingreliangxuefang (QRLXF) on psoriasis. Materials and Methods We used network pharmacology, a computational approach, to identify key bioactive compounds and biological targets, and explored the molecular mechanisms of QRLXF. The effects of QRLXF on keratinocyte proliferation and inflammation were evaluated using a mouse model of psoriasis. Changes in the gut microbiota were analyzed via 16SrDNA sequencing, and T cell subsets were assessed using flow cytometry. Results Network pharmacology analysis suggested that QRLXF ameliorated psoriasis by modulating Th17 cell differentiation. Further experiments confirmed the anti-inflammatory effects and relief of psoriatic lesions in IMQ-induced mice. 16SrDNA sequencing revealed significant shifts in the gut microbiota, notably increases in Ligilactobacillus and Lactobacillus genera and decreases in Anaerotruncus, Negativibacillus, Bilophila, and Mucispirillum, suggesting a potential relationship between specific microbiota changes and Th17 cell differentiation. Conclusion QRLXF alleviated psoriatic dermatitis by regulating Th17 cell responses and modifying gut microbiota profiles, highlighting its therapeutic potential for psoriasis treatment.
Collapse
Affiliation(s)
- Zhengyao Yu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Yongkang Hospital of Traditional Chinese Medicine, Jinhua, Zhejiang, 321300, People’s Republic of China
| | - Yingying Wang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Yingxue Guo
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Ruotong Zhu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Yimiao Fang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
| | - Qinghua Yao
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310005, People’s Republic of China
| | - Huiying Fu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Yongkang Hospital of Traditional Chinese Medicine, Jinhua, Zhejiang, 321300, People’s Republic of China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Ang Zhou
- Department of Dermatology, Yiwu Central Hospital Medical Community Choujiang Hospital District, Yiwu, Zhejiang, 322000, People’s Republic of China
| | - Lili Ma
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
| | - Qiyang Shou
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Yongkang Hospital of Traditional Chinese Medicine, Jinhua, Zhejiang, 321300, People’s Republic of China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| |
Collapse
|
27
|
Kolland D, Kuhlmann M, de Almeida GP, Köhler A, Arifovic A, von Strempel A, Pourjam M, Bolsega S, Wurmser C, Steiger K, Basic M, Neuhaus K, Schmidt-Weber CB, Stecher B, Zehn D, Ohnmacht C. A specific microbial consortium enhances Th1 immunity, improves LCMV viral clearance but aggravates LCMV disease pathology in mice. Nat Commun 2025; 16:3902. [PMID: 40274773 PMCID: PMC12022176 DOI: 10.1038/s41467-025-59073-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Anti-viral immunity can vary tremendously from individual to individual but mechanistic understanding is still scarce. Here, we show that a defined, low complex bacterial community (OMM12) but not the general absence of microbes in germ-free mice leads to a more potent immune response compared to the microbiome of specific-pathogen-free (SPF) mice after a systemic viral infection with LCMV Clone-13. Consequently, gnotobiotic mice colonized with OMM12 have more severe LCMV-induced disease pathology but also enhance viral clearance in the intestinal tract. Mechanistically, single-cell RNA sequencing analysis of adoptively transferred virus-specific T helper cells and endogenous T helper cells in the intestinal tract reveal a stronger pro-inflammatory Th1 profile and a more vigorous expansion in OMM12 than SPF mice. Altogether, our work highlights the causative function of the intestinal microbiome for shaping adaptive anti-viral immunity with implications for vaccination strategies and anti-cancer treatment regimens.
Collapse
Affiliation(s)
- Daphne Kolland
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Miriam Kuhlmann
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Gustavo P de Almeida
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Amelie Köhler
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Anela Arifovic
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Alexandra von Strempel
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU, Munich, Germany
| | - Mohsen Pourjam
- Core Facility Microbiome ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Silvia Bolsega
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Christine Wurmser
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technical University Munich, Munich, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Klaus Neuhaus
- Core Facility Microbiome ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
- Member of the German Center of Lung Research (DZL), Partner Site Munich, Munich, Germany
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU, Munich, Germany
- German Center for Infection Research (DZIF), partner site LMU, Munich, Germany
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany.
| |
Collapse
|
28
|
Shi S, Zhang J, Zhang J, Ma S, Hu Y, Zhu H, Wang H, Jiang M, Wang Y. Structural characterization of raw and wine-steamed Polygonatum cyrtonema Hua oligosaccharides and their bioactivity on immune regulation via modifying the gut microbiota. Int Immunopharmacol 2025; 153:114468. [PMID: 40154181 DOI: 10.1016/j.intimp.2025.114468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/16/2025] [Accepted: 03/09/2025] [Indexed: 04/01/2025]
Abstract
Polygonatum cyrtonema Hua (PC) is a traditional Chinese medicine with a long history of use as pharmaceuticals or functional foods. Wine steaming is the main processing method of PC, which changes the structure of polysaccharides and oligosaccharides in PC and enhances biological activities. This study investigated the structural characterization of raw and wine-steamed PC oligosaccharides and the differences in the immunomodulatory effects using cyclophosphamide (CTX)-induced immunosuppression rat model. The oligosaccharides content and molecular weight of PC after wine steaming decreased, the proportion of oligosaccharides in total sugars and the reducing sugars content increased, and the monosaccharides composition of oligosaccharides changed. The raw Polygonatum cyrtonema Hua oligosaccharides (PCCO) and the wine-steamed Polygonatum cyrtonema Hua oligosaccharides (PCWO) exerted regulatory effects on organ index, immunoglobulin G (IgG), complement 3 (C3) spleen and colon tissue morphology, hematopoietic function of immunosuppressive rats treated by cyclophosphamide (CTX). Both PCCO and PCWO significantly regulated and improved the diversity and abundance of gut microbiota in immunosuppressed rats and increased the content of short-chain fatty acids (SCFAs) in the feces of rats, and the regulating effect of PCWO was better than PCCO. Differential microbiota analysis showed that PCWO could promote the proliferation of Bifidobacterium, Bacteroides, Oscillibacter, Roseburia, and Alistipes. In summary, the difference in the structural characteristics of PC oligosaccharides might be the reason for immune enhancement. This study could provide a theoretical basis for clarifying the scientific connotation of wine steaming to enhance the efficacy of PC, and promote the application of wine-steamed PC as an immunomodulator in pharmaceuticals or functional foods.
Collapse
Affiliation(s)
- Shuanghui Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 102488 Beijing, China
| | - Jingqiu Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 102488 Beijing, China
| | - Junli Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 102488 Beijing, China
| | - Siyuan Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 102488 Beijing, China
| | - Yufeng Hu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 102488 Beijing, China
| | - Haiting Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 102488 Beijing, China
| | - Huinan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 102488 Beijing, China
| | - Mingrui Jiang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 102488 Beijing, China
| | - Yingzi Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 102488 Beijing, China.
| |
Collapse
|
29
|
Xie R, Guo Z, Gan H, Hou D, Chen G, Deng C, Li H, Ouyang J, Tian Q, Huang X. Effects of Phytosterols on Growth Performance, Serum Indexes, and Fecal Microbiota in Finishing Pigs. Animals (Basel) 2025; 15:1188. [PMID: 40362003 PMCID: PMC12070934 DOI: 10.3390/ani15091188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/02/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Phytosterols (PSs) are widely present in plants, particularly abundant in plant oils and seeds. PSs are reported to have various biological activities, such as lowering cholesterol, alongside antioxidant and antibacterial activities. This research examined the effects of PSs in finishing pigs, including growth performance, serum biochemistry, and fecal bacteria. Two treatment groups (each treatment group consisted of five biological replicates, and each replicate comprised five pigs housed communally) were randomly assigned to the fifty finishing pigs (equally divided by sex) of "Duroc × Landrace × Yorkshire" three-way cross with 79.76 ± 1.29 (kg) body weight. The control group (CON) was given basic food, while the experimental group was given basic food containing 300 mg PS/kg (PS). Dietary PS supplementation markedly raised the levels of average daily feed intake (ADFI) and apparent digestibility of dry matter (DM), crude protein (CP), ether extract (EE), and gross energy (GE) in comparison to the CON (p < 0.05). Additionally, PSs also significantly boosted the concentrations of high-density lipoprotein cholesterol (HDL-C), total protein (TP), catalase (CAT), superoxide dismutase (SOD), total antioxidant capacity (T-AOC), immunoglobulin G (IgG), motilin (MTL), and glucagon-like peptide-1 (GLP-1) (p < 0.05), and lowered the lactate dehydrogenase (LDH) level (p < 0.05). Both at the phyla and genus levels, the relative abundance of Firmicutes and Streptococcus increased significantly, and the relative abundance of Acinetobacter decreased significantly when adding phytosterols (p < 0.05). Overall, phytosterols dietary supplementation promotes immunity and antioxidant capacity in finishing pigs and boosts growth performance by improving nutrient digestibility.
Collapse
Affiliation(s)
- Renjie Xie
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (R.X.); (Z.G.)
- Yuelushan Laboratory, Changsha 410128, China
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| | - Zhenxing Guo
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (R.X.); (Z.G.)
| | - Haiqing Gan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (R.X.); (Z.G.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Dexing Hou
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| | - Guang Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (R.X.); (Z.G.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Chao Deng
- Hunan Biological and Electromechanical Polytechnic, Changsha 210127, China
| | - Hongkun Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (R.X.); (Z.G.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Jiajie Ouyang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (R.X.); (Z.G.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Qiyu Tian
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (R.X.); (Z.G.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Xingguo Huang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (R.X.); (Z.G.)
- Yuelushan Laboratory, Changsha 410128, China
| |
Collapse
|
30
|
Deng Y, Zhang Y, Xiao J, Cao Y, Ho CT, Lu M. Allicin Improves Diet-Induced Nonalcoholic Steatohepatitis and Gut Microbiota Dysbiosis in Mice via the Involvement of the Circadian Clock Gene Rev-erbα. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9019-9032. [PMID: 40168418 DOI: 10.1021/acs.jafc.4c12566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Nonalcoholic Steatohepatitis (NASH) is a progressive liver disease characterized by inflammation and liver damage. Allicin, a bioactive compound derived from garlic, has demonstrated anti-inflammatory and antioxidant properties. This study explores the effects of allicin on NASH and gut microbiota dysbiosis induced by a high-fat, high-fructose diet (HFFD) in mice. Allicin supplementation significantly alleviated hepatic inflammation, improved glucose metabolism, and modulated the circadian rhythm gene Rev-erbα, which plays a critical role in regulating inflammation. The anti-inflammatory effects of allicin were diminished in Si-Rev-erbα-treated HepG2 cells, highlighting the importance of circadian regulation in mediating these effects. Allicin's anti-inflammatory effects were associated with increased levels of short-chain fatty acids (SCFAs) and the restoration of diurnal oscillations in proinflammatory cytokines and gut microbiota composition, particularly in genera, such as Akkermansia, Bacteroidetes, and Lactobacillus. These findings suggest that allicin could be a promising therapeutic approach for managing NASH, liver dysfunction, and related metabolic disorders through the modulation of circadian rhythms and the gut microbiome.
Collapse
Affiliation(s)
- Yupei Deng
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yiyi Zhang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Muwen Lu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
31
|
El Boukhari R, Matin M, Bouissane L, Ławiński M, Lushchak O, Singla RK, Mickael M, Mayneris‐Perxachs J, Grafakou ME, Xu S, Liu B, Guan J, Półtorak A, Szpicer A, Wierzbicka A, Tzvetkov NT, Banach M, Horbańczuk JO, Jóźwik A, Cascella M, Shen B, Pirgozliev VR, Wang D, Litvinova O, Adamska O, Kamińska A, Łapiński M, Stolarczyk A, Berindan‐Neagoe I, Milella L, Yeung AWK, Suravajhala P, Bishayee A, Lordan R, Iantovics LB, Lagoa R, Michalczuk M, Stoyanov J, Kinghorn AD, Jalil B, Weckwerth W, Goh BH, Li M, Chaubey G, Russo GL, Frazzini S, Rossi L, Battino M, Jia W, Su Q, Ma X, Rollinger JM, Rittmann SKR, Sheridan H, Walsh JJ, Lizard G, Karpiński TM, Silva AS, Piwowarski J, Xie L, Fan T, Giampieri F, El Midaoui A, Wong K, Gan R, Fatimi A, Atanasov AG. Enhancing human gut health: Global innovations in dysbiosis management. IMETA 2025. [DOI: 10.1002/imt2.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/26/2025] [Indexed: 05/03/2025]
Affiliation(s)
- Reda El Boukhari
- Chemical Science and Engineering Research Team (ERSIC), Department of Chemistry, Polydisciplinary Faculty of Beni Mellal (FPBM) Sultan Moulay Slimane University (USMS) Beni Mellal Morocco
| | - Maima Matin
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Latifa Bouissane
- Molecular Chemistry, Materials and Catalysis Laboratory, Faculty of Sciences and Technologies Sultan Moulay Slimane University Beni Mellal Morocco
| | - Michał Ławiński
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
- Department of General, Gastroenterologic and Oncologic Surgery Medical University of Warsaw Warsaw Poland
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology Vasyl Stefanyk Precarpathian National University Ivano‐Frankivsk Ukraine
- Research and Development University Ivano‐Frankivsk Ukraine
| | - Rajeev K. Singla
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu Sichuan China
- School of Pharmaceutical Sciences Lovely Professional University Phagwara Punjab India
| | - Michel‐Edwar Mickael
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Jordi Mayneris‐Perxachs
- Department of Diabetes, Endocrinology and Nutrition Dr. Josep Trueta University Hospital Girona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) Madrid Spain
- Integrative Systems Medicine and Biology Group, Girona Biomedical Research Institute (IDIBGI‐CERCA) Parc Hospitalari Martí i Julià Salt Spain
| | - Maria Eleni Grafakou
- Chair of Pharmaceutical Biology, Faculty of Pharmacy and Chemistry University of Regensburg Germany
| | - Shuhua Xu
- Center for Evolutionary Biology, School of Life Sciences Fudan University Shanghai China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center Fudan University Shanghai China
| | - Bowen Liu
- School of Agriculture Yunnan University Kunming China
| | - Jiayi Guan
- Henan Institute of Medical and Pharmaceutical Sciences Zhengzhou University Zhengzhou China
| | - Andrzej Półtorak
- Department of Technique and Food Development, Institute of Human Nutrition Sciences Warsaw University of Life Sciences Warsaw Poland
| | - Arkadiusz Szpicer
- Department of Technique and Food Development, Institute of Human Nutrition Sciences Warsaw University of Life Sciences Warsaw Poland
| | - Agnieszka Wierzbicka
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
- Department of Technique and Food Development, Institute of Human Nutrition Sciences Warsaw University of Life Sciences Warsaw Poland
| | - Nikolay T. Tzvetkov
- Department of Biochemical Pharmacology and Drug Design, Institute of Molecular Biology “Roumen Tsanev” Bulgarian Academy of Sciences Sofia Bulgaria
| | - Maciej Banach
- Faculty of Medicine The John Paul II Catholic University of Lublin (KUL) Lublin Poland
- Department of Cardiology and Adult Congenital Heart Diseases Polish Mother's Memorial Hospital Research Institute (PMMHRI) Lodz Poland
- Department of Preventive Cardiology and Lipidology Medical University of Lodz (MUL) Lodz Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Jarosław Olav Horbańczuk
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Artur Jóźwik
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Marco Cascella
- Anesthesia and Pain Medicine, Department of Medicine, Surgery and Dentistry “Scuola MedicaSalernitana” University of Salerno Baronissi Italy
| | - Bairong Shen
- Department of Critical Care Medicine and Institutes for Systems Genetics Frontiers Science Center for Disease‐Related Molecular Network, West China Hospital,Sichuan University Chengdu Sichuan China
- Center for High Altitude Medicine, West China Hospital Sichuan University Chengdu Sichuan China
| | | | - Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research McMaster University Hamilton Ontario Canada
- Division of Endocrinology and Metabolism, Department of Medicine McMaster University Hamilton Ontario Canada
| | - Olena Litvinova
- National University of Pharmacy of the Ministry of Health of Ukraine Kharkiv Ukraine
- Ludwig Boltzmann Institute Digital Health and Patient Safety Medical University of Vienna Vienna Austria
| | - Olga Adamska
- Faculty of Medicine Collegium Medicum Cardinal Stefan Wyszyński University in Warsaw Warsaw Poland
| | - Agnieszka Kamińska
- Faculty of Medicine Collegium Medicum Cardinal Stefan Wyszyński University in Warsaw Warsaw Poland
| | - Marcin Łapiński
- Orthopaedic and Rehabilitation Department Medical University of Warsaw Warsaw Poland
| | - Artur Stolarczyk
- Orthopaedic and Rehabilitation Department Medical University of Warsaw Warsaw Poland
| | - Ioana Berindan‐Neagoe
- Department of Genomics MEDFUTURE ‐ Institute for Biomedical Research“Iuliu Hațieganu” University of Medicine and Pharmacy No. 23 Cluj‐Napoca Romania
| | - Luigi Milella
- Department of Health Sciences University of Basilicata Potenza Italy
| | - Andy Wai Kan Yeung
- Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry The University of Hong Kong Pokfulam Hong Kong SAR
| | - Prashanth Suravajhala
- Amrita School of Biotechnology Amrita Viswa Vidyapeetham Clappana Kerala India
- Department of Biosciences Manipal University Jaipur, Dehmi Kala Jaipur Rajasthan India
| | - Anupam Bishayee
- Department of Pharmacology College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine Bradenton Florida USA
| | - Ronan Lordan
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| | - Laszlo Barna Iantovics
- Department of Electrical Engineering and Information Technology George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures Targu Mures Romania
| | - Ricardo Lagoa
- ESTG‐Polytechnic Institute of Leiria Morro do Lena‐Alto do Vieiro Leiria Portugal
- LSRE‐LCM‐Associate Laboratory in Chemical Engineering University of Porto Porto Portugal
| | - Monika Michalczuk
- Department of Animal Breeding, Institute of Animal Sciences Warsaw University of Life Sciences ‐ SGGW Warsaw Poland
| | - Jivko Stoyanov
- Swiss Paraplegic Research Nottwil Switzerland
- Institute of Social and Preventive Medicine (ISPM) University of Bern Bern Switzerland
| | | | - Banaz Jalil
- Pharmacognosy and Phytotherapy UCL School of Pharmacy London UK
| | - Wolfram Weckwerth
- Molecular Systems Biology Lab (MOSYS), Department of Functional and Evolutionary Ecology University of Vienna Vienna Austria
- Vienna Metabolomics Center (VIME) University of Vienna Vienna Austria
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre (SBMDC) School of Medical and Life Sciences Subang Jaya Malaysia
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy Monash University Malaysia Subang Jaya Malaysia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine University of Technology Sydney Ultimo New South Wales Australia
| | - Meng‐Yao Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
- Department of Biliary‐Pancreatic Surgery, Renji Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Gyaneshwer Chaubey
- Cytogenetics Laboratory, Department of Zoology Banaras Hindu University Varanasi Uttar Pradesh India
| | - Gian Luigi Russo
- National Research Council Institute of Food Sciences Avellino Italy
| | - Sara Frazzini
- Department of Veterinary Medicine and Animal Science (DIVAS) University of Milan Lodi Italy
| | - Luciana Rossi
- Department of Veterinary Medicine and Animal Science (DIVAS) University of Milan Lodi Italy
| | - Maurizio Battino
- Department of Clinical Sciences Polytechnic University of Marche Ancona Italy
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods Polytechnic University of Marche (Italy), Universidad Europea del Atlántico (Spain), and Jiangsu University (China) Ancona Italy
- International Joint Research, Laboratory of Intelligent Agriculture and Agri‐Products Processing Jiangsu University Zhenjiang China
| | - Wei Jia
- Department of Pharmacology and Pharmacy The University of Hong Kong Pokfulam Hong Kong SAR
| | - Qi Su
- Microbiota I‐Center Shatin Hong Kong SAR
- Department of Medicine and Therapeutics The Chinese University of Hong Kong Shatin Hong Kong SAR
| | - Xiaoqiang Ma
- Department of Food Science and Technology, School of Agriculture and Biology Shanghai Jiao Tong University Shanghai China
| | - Judith M. Rollinger
- Division of Pharmacognosy, Department of Pharmaceutical Sciences, Faculty of Life Sciences University of Vienna Vienna Austria
| | - Simon K.‐M. R. Rittmann
- Archaea Physiology & Biotechnology Group, Department of Functional and Evolutionary Ecology University of Vienna Vienna Austria
| | - Helen Sheridan
- The NatPro Centre & School of Pharmacy and Pharmaceutical Sciences Trinity College Dublin Dublin Ireland
- Université Bourgogne Europe/INSERM, 21000 Dijon and PHYNOHA Consulting Fontaine‐lès‐Dijon France
| | - John J. Walsh
- The NatPro Centre & School of Pharmacy and Pharmaceutical Sciences Trinity College Dublin Dublin Ireland
| | - Gérard Lizard
- Université Bourgogne Europe/INSERM, 21000 Dijon and PHYNOHA Consulting Fontaine‐lès‐Dijon France
| | - Tomasz M. Karpiński
- Department of Medical Microbiology Poznań University of Medical Sciences Poznań Poland
| | - Ana Sanches Silva
- University of Coimbra, Faculty of Pharmacy, Polo III, Azinhaga de Santa Comba Coimbra Portugal
- Centre for Animal Science Studies (CECA), ICETA University of Porto Porto Portugal
| | - Jakub Piwowarski
- Microbiota Lab, Department of Pharmaceutical Microbiology and Bioanalysis Medical University of Warsaw Warsaw Poland
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology Institute of Microbiology, Guangdong Academy of Sciences Guangzhou China
- School of Life & Health Sciences Fuyao University of Science & Technology Fuzhou Fujian China
| | - Tai‐Ping Fan
- School of Life & Health Sciences Fuyao University of Science & Technology Fuzhou Fujian China
| | - Francesca Giampieri
- Department of Clinical Sciences Polytechnic University of Marche Ancona Italy
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods Polytechnic University of Marche (Italy), Universidad Europea del Atlántico (Spain), and Jiangsu University (China) Ancona Italy
- Research Group on Food, Nutritional Biochemistry and Health Universidad Europea del Atlántico Santander Spain
- International Research Center for Food Nutrition and Safety Jiangsu University Zhenjiang China
| | - Adil El Midaoui
- Faculty of Sciences and Techniques Errachidia, Moulay Ismail University of Meknes Meknes Morocco
- Department of Pharmacology and Physiology, Faculty of Medicine University of Montreal Montreal Quebec Canada
| | - Ka‐Hing Wong
- Research Institute for Future Food The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
- Department of Food Science and Nutrition The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
| | - Ren‐You Gan
- Research Institute for Future Food The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
- Department of Food Science and Nutrition The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
| | - Ahmed Fatimi
- Chemical Science and Engineering Research Team (ERSIC), Department of Chemistry, Polydisciplinary Faculty of Beni Mellal (FPBM) Sultan Moulay Slimane University (USMS) Beni Mellal Morocco
| | - Atanas G. Atanasov
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
- Ludwig Boltzmann Institute Digital Health and Patient Safety Medical University of Vienna Vienna Austria
- Laboratory of Natural Products and Medicinal Chemistry (LNPMC), Center for Global Health Research, Saveetha Medical College and Hospital Saveetha Institute of Medical and Technical Sciences (SIMATS) Thandalam Chennai India
| |
Collapse
|
32
|
Han J, Ren Y, Zhang P, Fang C, Yang L, Zhou S, Ji Z. The effectiveness of treatment with probiotics in preventing necrotizing enterocolitis and related mortality: results from an umbrella meta-analysis on meta-analyses of randomized controlled trials. BMC Gastroenterol 2025; 25:245. [PMID: 40217146 PMCID: PMC11987312 DOI: 10.1186/s12876-025-03788-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
INTRODUCTION Probiotic supplementation has been proposed as a preventive measure for necrotizing enterocolitis (NEC) in preterm infants. This umbrella meta-analysis assesses the effects of probiotics, including single-strain and multi-strain formulations, on NEC and related mortality. METHODS A comprehensive search was conducted in PubMed, Scopus, ISI Web of Science, and Embase for studies up to August 2024. The AMSTAR2 tool assessed the quality of included studies. Meta-analysis studies were selected based on the PICOS framework, focusing on preterm neonates (< 37-week gestation), probiotic supplementation (single-strain or multi-strain), placebo or standard care comparison, and outcomes of NEC and mortality. Pooled relative risks (RR) and odds ratios (OR) with 95% confidence intervals (CI) were calculated using random-effects models. RESULTS Overall, 35 eligible studies were included into the study. Twenty-six and 32 probiotic intervention arms used single- and multi-strain probiotics, respectively. The findings revealed that probiotics decreased NEC significantly (ESRR: 0.51; 95% CI: 0.46, 0.55, p < 0.001, and ESOR: 0.59; 95%CI: 0.48, 0.72, P < 0.001), and mortality rate (ESRR: 0.72; 95% CI: 0.68, 0.76, P < 0.001, and ESOR: 0.77; 95%CI: 0.70, 0.84, p < 0.001). CONCLUSION The present review suggests that supplementation with probiotics reduced NEC and related mortality. Probiotic supplementation can be recognized as a NEC-preventing approach in preterm and very preterm infants, particularly Multi-strain probiotics.
Collapse
Affiliation(s)
- Jiaju Han
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Yufeng Ren
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China.
| | - Peini Zhang
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Chengfeng Fang
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Leilei Yang
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Shenkang Zhou
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Zhiqing Ji
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| |
Collapse
|
33
|
Eslami M, Adampour Z, Fadaee Dowlat B, Yaghmayee S, Motallebi Tabaei F, Oksenych V, Naderian R. A Novel Frontier in Gut-Brain Axis Research: The Transplantation of Fecal Microbiota in Neurodegenerative Disorders. Biomedicines 2025; 13:915. [PMID: 40299512 PMCID: PMC12025253 DOI: 10.3390/biomedicines13040915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
The gut-brain axis (GBA) represents a sophisticated bidirectional communication system connecting the central nervous system (CNS) and the gastrointestinal (GI) tract. This interplay occurs primarily through neuronal, immune, and metabolic pathways. Dysbiosis in gut microbiota has been associated with multiple neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS). In recent years, fecal microbiota transplantation (FMT) has gained attention as an innovative therapeutic approach, aiming to restore microbial balance in the gut while influencing neuroinflammatory and neurodegenerative pathways. This review explores the mechanisms by which FMT impacts the gut-brain axis. Key areas of focus include its ability to reduce neuroinflammation, strengthen gut barrier integrity, regulate neurotransmitter production, and reinstate microbial diversity. Both preclinical and clinical studies indicate that FMT can alleviate motor and cognitive deficits in PD and AD, lower neuroinflammatory markers in MS, and enhance respiratory and neuromuscular functions in ALS. Despite these findings, several challenges remain, including donor selection complexities, uncertainties about long-term safety, and inconsistencies in clinical outcomes. Innovations such as synthetic microbial communities, engineered probiotics, and AI-driven analysis of the microbiome hold the potential to improve the precision and effectiveness of FMT in managing neurodegenerative conditions. Although FMT presents considerable promise as a therapeutic development, its widespread application for neurodegenerative diseases requires thorough validation through well-designed, large-scale clinical trials. It is essential to establish standardized protocols, refine donor selection processes, and deepen our understanding of the molecular mechanisms behind its efficacy.
Collapse
Affiliation(s)
- Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran;
- Department of Bacteriology and Virology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Zarifeh Adampour
- Institute of Science, Biotechnology and Biosafety Department, Eskishehir Osmangazi University, Eskishehir 26040, Türkiye;
| | - Bahram Fadaee Dowlat
- School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Shayan Yaghmayee
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Faezeh Motallebi Tabaei
- Department of Medical Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 49189-36316, Iran
| | | | - Ramtin Naderian
- Clinical Research Development Unit, Kowsar Educational, Research and Therapeutic Hospital, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| |
Collapse
|
34
|
Osaki M, Sakaguchi S. Soluble CTLA-4 regulates immune homeostasis and promotes resolution of inflammation by suppressing type 1 but allowing type 2 immunity. Immunity 2025; 58:889-908.e13. [PMID: 40168991 DOI: 10.1016/j.immuni.2025.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/29/2024] [Accepted: 03/05/2025] [Indexed: 04/03/2025]
Abstract
Cytotoxic T-lymphocyte-associated antigen -4 (CTLA-4) is a co-inhibitory receptor that restricts T cell activation. CTLA-4 exists as membrane (mCTLA-4) and soluble (sCTLA-4) forms, but the key producers, kinetics, and functions of sCTLA-4 are unclear. Here, we investigated the roles of sCTLA-4 in immune regulation under non-inflammatory and inflammatory conditions. Effector regulatory T (Treg) cells were the most active sCTLA-4 producers in basal and inflammatory states, with distinct kinetics upon T cell receptor (TCR) stimulation. We generated mice specifically deficient in sCTLA-4 production, which exhibited spontaneous activation of type 1 immune cells and heightened autoantibody/immunoglobulin E (IgE) production. Conversely, mCTLA-4-deficient mice developed severe type 2-skewed autoimmunity. sCTLA-4 blockade of CD80/86 on antigen-presenting cells inhibited T helper (Th)1, but not Th2, differentiation in vitro. In vivo, Treg-produced sCTLA-4, suppressed Th1-mediated experimental colitis, and enhanced wound healing but hampered tumor immunity. Thus, sCTLA-4 is essential for immune homeostasis and controlling type 1 immunity while allowing type 2 immunity to facilitate resolution in inflammatory conditions.
Collapse
Affiliation(s)
- Motonao Osaki
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan; Laboratory of Experimental Immunology, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan; Laboratory of Experimental Immunology, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
35
|
Cao J, Wang S, Ding R, Liu Y, Yuan B. Comparative analyses of the gut microbiome of two sympatric rodent species, Myodes rufocanus and Apodemus peninsulae, in northeast China based on metagenome sequencing. PeerJ 2025; 13:e19260. [PMID: 40226542 PMCID: PMC11988107 DOI: 10.7717/peerj.19260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
The gut microbiota is integral to an animal's physiology, influencing nutritional metabolism, immune function, and environmental adaptation. Despite the significance of gut microbiota in wild rodents, the Korean field mouse (Apodemus peninsulae) and the gray red-backed vole (Myodes rufocanus) remain understudied. To address this, a metagenomic sequencing analysis of the gut microbiome of these sympatric rodents in northeast China's temperate forests was conducted. Intestinal contents were collected from A. peninsulae and M. rufocanus within the Mudanfeng National Nature Reserve. High-throughput sequencing elucidated the gut microbiome's composition, diversity, and functional pathways. Firmicutes, Bacteroidetes, and Proteobacteria were identified as the dominant phyla, with M. rufocanus showing greater microbiome diversity. Key findings indicated distinct gut bacterial communities between the species, with M. rufocanus having a higher abundance of Proteobacteria. The gut microbiota of A. peninsulae and M. rufocanus differed marginally in functional profiles, specifically in the breakdown of complex carbohydrates, which might reflect their distinct food preferences albeit both being herbivores with a substantial dietary overlap. The investigation further elucidated gut microbiota's contributions to energy metabolism and environmental adaptation mechanisms. This study aligns with information on rodent gut microbiota in literature and highlights the two understudied rodent species, providing comparative data for future studies investigating the role of gut microbiota in wildlife health and ecosystem functioning.
Collapse
Affiliation(s)
- Jing Cao
- College of Biology and Food, Shangqiu Normal University, Shangqiu, Henan, China
| | - Shengze Wang
- School of Life Science, Liaocheng University, Liaocheng, Shandong, China
| | - Ruobing Ding
- College of Biology and Food, Shangqiu Normal University, Shangqiu, Henan, China
| | - Yijia Liu
- College of Biology and Food, Shangqiu Normal University, Shangqiu, Henan, China
| | - Baodong Yuan
- School of Life Science, Liaocheng University, Liaocheng, Shandong, China
| |
Collapse
|
36
|
Yuan X, Wang J, Wang W, Song Y, Wu J, Du R. Microbiome alterations in primary Sjögren's syndrome: Regional dysbiosis and microbiome-targeted therapeutic strategies. Clin Immunol 2025; 273:110444. [PMID: 39947272 DOI: 10.1016/j.clim.2025.110444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/27/2025] [Accepted: 02/08/2025] [Indexed: 02/21/2025]
Abstract
Primary Sjögren's syndrome (pSS) is a complex autoimmune disease characterized by diverse clinical manifestations. While xerophthalmia and xerostomia are hallmark symptoms, the disease often involves multiple organ systems, including the kidneys, lungs, nervous system, and gastrointestinal tract, leading to systemic morbidity in severe cases. Despite extensive research, the precise pathogenesis of pSS remains unclear, likely involving infectious, hormonal, and genetic factors. Emerging evidence highlights the microbiome as a key contributor to autoimmune diseases, including pSS. Dysbiosis in the oral, ocular, gut, and genital microbiomes plays a critical role in disease onset, progression, and variability. This review summarizes current findings on microbiome alterations in pSS, emphasizing their role in pathogenesis and clinical features, and explores microbiome-targeted therapies. Understanding the role of the microbiome in pSS pathophysiology could advance disease management and inspire targeted therapeutic strategies.
Collapse
Affiliation(s)
- Xujing Yuan
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jun Wang
- Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weiwei Wang
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - You Song
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jiajia Wu
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Rong Du
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
37
|
Sakinah EN, Diniyah N, Subagio A, Marchianti ACN, Firdaus J, Fambudi SS, Amini SA, Putri DE. Exploring the role of polysaccharides in mitigating organ damage caused by pesticide-induced toxicity: A systematic review and meta-analysis of in vivo studies. NARRA J 2025; 5:e1553. [PMID: 40352195 PMCID: PMC12059844 DOI: 10.52225/narra.v5i1.1553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/13/2025] [Indexed: 05/14/2025]
Abstract
Although polysaccharides have demonstrated potential in alleviating dysbiosis, the overall impact of polysaccharides on minimizing oxidative stress and organ damage in vivo has not been thoroughly investigated. The aim of this study was to investigate the comprehensive effects of polysaccharides in mitigating pesticide toxicity in animal studies, focusing on biomarkers related to oxidative stress, antioxidant activity, kidney injury, lipid profiles, liver function, and the preservation of liver and kidney weights. A systematic search was conducted across nine indexed databases, including PubMed, Cochrane CENTRAL, Taylor & Francis, Scopus, Sage, EBSCO, ProQuest, ScienceDirect, and Google Scholar. Rayyan.ai was used to screen in vivo studies that met the predefined inclusion and exclusion criteria. The quality of the selected in vivo studies was evaluated using SYRCLE's Risk of Bias tool, specifically designed for animal studies. Thirteen randomized animal studies, comprising 330 mice and rats, were included in the analysis. The findings revealed that polysaccharides significantly increased antioxidant levels, including catalase (CAT) (p<0.00001), superoxide dismutase (SOD) (p<0.00001), glutathione peroxidase (GPx) (p<0.00001), and reduced glutathione (GSH) (p<0.00001). Polysaccharides also significantly reduced oxidative stress markers, such as malondialdehyde (MDA) (p<0.00001) and nitric oxide (NO) (p<0.0001), as well as kidney injury biomarkers, including serum creatinine (p<0.00001) and urea (p<0.00001). Additionally, improvements in lipid profiles were observed, with significant reductions in triglycerides (TG) (p=0.04) and total cholesterol (TC) (p<0.00001). However, there were no significant differences in high-density lipoprotein (HDL) (p=0.28) and low-density lipoprotein (LDL) (p=0.32) levels. Polysaccharides significantly alleviate liver biomarkers, including aspartate transaminase (AST) (p<0.0001), alanine transaminase (ALT) (p<0.005), and alkaline phosphatase (ALP) (p<0.0001). Polysaccharides also contributed to the maintenance of liver weight (p=0.009), although no significant differences were observed in kidney weights (p=0.81). The study highlights that polysaccharides exert significant effects in enhancing antioxidant levels, reducing oxidative stress and organ damage biomarkers, and preserving liver weights.
Collapse
Affiliation(s)
- Elly N. Sakinah
- Department of Pharmacology, Faculty of Medicine, Universitas Jember, Jember, Indonesia
| | - Nurud Diniyah
- Department of Agricultural Products Technology, Faculty of Agricultural Technology, Universitas Jember, Jember, Indonesia
| | - Achmad Subagio
- Department of Agricultural Products Technology, Faculty of Agricultural Technology, Universitas Jember, Jember, Indonesia
| | - Ancah CN. Marchianti
- Department of Public Health, Faculty of Medicine, Universitas of Jember, Jember, Indonesia
| | - Jauhar Firdaus
- Department of Physiology, Faculty of Medicine, Universitas Jember, Jember, Indonesia
| | | | | | | |
Collapse
|
38
|
Saadawi A, Mair F, Rosenwald E, Hoces D, Slack E, Kopf M. Investigating Polyreactivity of CD4 + T Cells to the Intestinal Microbiota. Eur J Immunol 2025; 55:e202451484. [PMID: 40223653 DOI: 10.1002/eji.202451484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/15/2025]
Abstract
Antigen-specific recognition of microbiota by T cells enforces tolerance at homeostasis. Conversely, dysbiosis leads to imbalanced T-cell responses, triggering inflammatory and autoimmune diseases. Despite their significance, the identities of immunogenic microbial antigens remain largely enigmatic. Here, we leveraged a sensitive, unbiased, genome-wide screening platform to identify peptides from Akkermansia muciniphila (AKK) and Bacteroides thetaiotaomicron (BT) recognized by CD4+ T cells. The platform is based on screening peptide libraries using an NFAT-fluorescence reporter cell line transduced with a retrovirus encoding an MHC-TCR (MCR) hybrid molecule. We discovered several novel epitopes from AKK and BT. T-cell hybridomas reactive to AKK and BT bacteria demonstrated polyreactivity to microbiota-derived peptides in co-cultures with MCR reporter cells. Steady-state T cells recognized these epitopes in an MHC-restricted fashion. Intriguingly, most of the identified epitopes are broadly conserved within the given phylum and originate from membrane and intracellular proteins. Ex vivo stimulation of CD4+ T cells from mice vaccinated with the identified peptides revealed mono-specific IFN-γ and IL-17 responses. Our work showcases the potential of the MCR system for identifying immunogenic microbial epitopes, providing a valuable resource. Our study facilitates decoding antigen specificity in immune system-bacterial interactions, with applications in understanding microbiome and pathogenic bacterial immunity.
Collapse
Affiliation(s)
- Ahmed Saadawi
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Florian Mair
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Esther Rosenwald
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Daniel Hoces
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Emma Slack
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Manfred Kopf
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
39
|
Schütz B, Krause FF, Taudte RV, Zaiss MM, Luu M, Visekruna A. Modulation of Host Immunity by Microbiome-Derived Indole-3-Propionic Acid and Other Bacterial Metabolites. Eur J Immunol 2025; 55:e202451594. [PMID: 40170399 PMCID: PMC11962249 DOI: 10.1002/eji.202451594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/03/2025]
Abstract
In recent years, we have witnessed a rapidly growing interest in the intricate communications between intestinal microorganisms and the host immune system. Research on the human microbiome is evolving from merely descriptive and correlative studies to a deeper mechanistic understanding of the bidirectional interactions between gut microbiota and the mucosal immune system. Despite numerous challenges, it has become increasingly evident that an imbalance in gut microbiota composition, known as dysbiosis, is associated with the development and progression of various metabolic, immune, cancer, and neurodegenerative disorders. A growing body of evidence highlights the importance of small molecules produced by intestinal commensal bacteria, collectively referred to as gut microbial metabolites. These metabolites serve as crucial diffusible messengers, translating the microbial language to host cells. This review aims to explore the complex and not yet fully understood molecular mechanisms through which microbiota-derived metabolites influence the activity of the immune cells and shape immune reactions in the gut and other organs. Specifically, we will discuss recent research that reveals the close relationship between microbial indole-3-propionic acid (IPA) and mucosal immunity. Furthermore, we will emphasize the beneficial effects of IPA on intestinal inflammation and discuss its potential clinical implications.
Collapse
Affiliation(s)
- Burkhard Schütz
- Institute of Anatomy and Cell BiologyPhilipps‐University MarburgMarburgGermany
| | - Felix F. Krause
- Institute for Medical Microbiology and HygienePhilipps‐University MarburgMarburgGermany
| | - R. Verena Taudte
- Core Facility for MetabolomicsDepartment of MedicinePhilipps‐University MarburgMarburgGermany
| | - Mario M. Zaiss
- Department of Internal Medicine 3Rheumatology and ImmunologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU) and Universitätsklinikum ErlangenErlangenGermany
- Deutsches Zentrum Immuntherapie (DZI)Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU) and Universitätsklinikum ErlangenErlangenGermany
| | - Maik Luu
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik IIUniversitätsklinikum WürzburgWürzburgGermany
| | - Alexander Visekruna
- Institute for Medical Microbiology and HygienePhilipps‐University MarburgMarburgGermany
| |
Collapse
|
40
|
Zugman M, Wong M, Jaime-Casas S, Pal SK. The gut microbiome and dietary metabolites in the treatment of renal cell carcinoma. Urol Oncol 2025; 43:244-253. [PMID: 39095306 DOI: 10.1016/j.urolonc.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/13/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024]
Abstract
The gut microbiome is interlinked with renal cell carcinoma (RCC) and its response to systemic treatment. Mounting data suggests that certain elements of the gut microbiome may correlate with improved outcomes. New generation sequencing techniques and advanced bioinformatic data curation are accelerating the investigation of specific markers and metabolites that could predict treatment response. A variety of new therapeutic strategies, such as fecal microbiota transplantation, probiotic supplements, and dietary interventions, are currently being developed to modify the gut microbiome and improve anticancer therapies in patients with RCC. This review discusses the preliminary evidence indicating the role of the microbiome in cancer treatment, the techniques and tools necessary for its proper study and some of the current forms with which the microbiome can be modulated to improve patient outcomes.
Collapse
Affiliation(s)
- Miguel Zugman
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA; Centro de Oncologia e Hematologia Família Dayan-Daycoval Einstein, Hospital Israelita Albert, São Paulo, São Paulo, Brazil
| | - Megan Wong
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Salvador Jaime-Casas
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Sumanta K Pal
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA.
| |
Collapse
|
41
|
Edwards M, Brockmann L. Microbiota-dependent modulation of intestinal anti-inflammatory CD4 + T cell responses. Semin Immunopathol 2025; 47:23. [PMID: 40167791 DOI: 10.1007/s00281-025-01049-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/08/2025] [Indexed: 04/02/2025]
Abstract
Barrier organs such as the gastrointestinal tract, lungs, and skin are colonized by diverse microbial strains, including bacteria, viruses, and fungi. These microorganisms, collectively known as the commensal microbiota, play critical roles in maintaining health by defending against pathogens, metabolizing nutrients, and providing essential metabolites. In the gut, commensal-derived antigens are frequently sensed by the intestinal immune system. Maintaining tolerance toward these beneficial microbial species is crucial, as failure to do so can lead to chronic inflammatory conditions like inflammatory bowel disease (IBD) and can even affect systemic immune or metabolic health. The immune system carefully regulates responses to commensals through various mechanisms, including the induction of anti-inflammatory CD4⁺ T cell responses. Foxp3⁺ regulatory T cells (Foxp3+ Tregs) and Type 1 regulatory T cells (Tr1) play a major role in promoting tolerance, as both cell types can produce the anti-inflammatory cytokine IL-10. In addition to these regulatory T cells, effector T cell subsets, such as Th17 cells, also adopt anti-inflammatory functions within the intestine in response to the microbiota. This process of anti-inflammatory CD4+ T cell induction is heavily influenced by the microbiota and their metabolites. Microbial metabolites affect intestinal epithelial cells, promoting the secretion of anti-inflammatory mediators that create a tolerogenic environment. They also modulate intestinal dendritic cells (DCs) and macrophages, inducing a tolerogenic state, and can interact directly with T cells to drive anti-inflammatory CD4⁺ T cell functionality. The disrupted balance of these signals may result in chronic inflammation, with broader implications for systemic health. In this review, we highlight the intricate interplays between commensal microorganisms and the immune system in the gut. We discuss how the microbiota influences the differentiation of commensal-specific anti-inflammatory CD4⁺ T cells, such as Foxp3⁺ Tregs, Tr1 cells, and Th17 cells, and explore the mechanisms through which microbial metabolites modulate these processes. We further discuss the innate signals that prime and commit these cells to an anti-inflammatory fate.
Collapse
Affiliation(s)
- Madeline Edwards
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Leonie Brockmann
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, 108-8345, Japan.
| |
Collapse
|
42
|
Wang Y, Waters AK, Basalirwa G, Ssetaala A, Mpendo J, Namuniina A, Keneema E, Kiiza D, Kyosiimire-Lugemwa J, Mayanja Y, Okech B, Kiwuwa-Muyingo S. Impact of Schistosoma mansoni Infection on the Gut Microbiome and Hepatitis B Vaccine Immune Response in Fishing Communities of Lake Victoria, Uganda. Vaccines (Basel) 2025; 13:375. [PMID: 40333219 PMCID: PMC12030974 DOI: 10.3390/vaccines13040375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 05/09/2025] Open
Abstract
OBJECTIVE Schistosoma mansoni (S. mansoni) infection is endemic in Ugandan fishing communities. We investigated its potential impact on Hepatitis B (Hep B) vaccine responses and its role in mediating the association between the gut microbiome and long-term effectiveness of the vaccine. METHODS Participants were tested for S. mansoni infections at baseline and received the Hep B vaccine at baseline, month 1, and month 6. Those with infections were treated. Stool samples were collected at baseline and analyzed using 16S rRNA sequencing. The Wilcoxon rank-sum test was used to compare alpha diversity between groups. A linear regression model was applied to estimate the association between one-year Hep B vaccine responses and the baseline gut microbiome by infection status, adjusting for age and sex. RESULTS A total of 107 participants were included (44 from the fishing community and 63 from the Kampala community). There was no significant difference in microbiome composition by location or infection status at baseline or discharge. In the linear regression analysis, S. mansoni infection (β = 1.24, p = 0.025) and a higher alpha diversity (β = 0.001, p = 0.07) were associated with higher Hep B vaccine responses, while older age was associated with a lower Hep B vaccine response (β = -0.06, p = 0.0013). CONCLUSIONS S. mansoni infection status before vaccination may modify the association between the gut microbiome and Hep B vaccine response. Potential interventions could focus on infection control as well as improving microbiome richness before implementing vaccine programs in fishing communities.
Collapse
Affiliation(s)
- Yan Wang
- Public and Population Health, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Ariana K. Waters
- School of Public Health, University of California, Berkeley, CA 94704, USA
| | | | - Ali Ssetaala
- UVRI-IAVI HIV Vaccine Program, Entebbe P.O. Box 49, Uganda
| | - Juliet Mpendo
- UVRI-IAVI HIV Vaccine Program, Entebbe P.O. Box 49, Uganda
| | | | - Emily Keneema
- UVRI-IAVI HIV Vaccine Program, Entebbe P.O. Box 49, Uganda
| | - David Kiiza
- UVRI-IAVI HIV Vaccine Program, Entebbe P.O. Box 49, Uganda
| | - Jacqueline Kyosiimire-Lugemwa
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM) Uganda Research Unit, P.O. Box 49, Plot 51-59 Nakiwogo Road, Entebbe 256, Uganda
- University of Kisubi, Faculty of Health Science and Department of Biomedical Sciences, Entebbe P.O. Box 182, Uganda
| | - Yunia Mayanja
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM) Uganda Research Unit, P.O. Box 49, Plot 51-59 Nakiwogo Road, Entebbe 256, Uganda
| | - Brenda Okech
- UVRI-IAVI HIV Vaccine Program, Entebbe P.O. Box 49, Uganda
| | - Sylvia Kiwuwa-Muyingo
- African Population & Health Research Center (APHRC), Nairobi Kenya P.O. Box 10787-00100, Kenya
| |
Collapse
|
43
|
Fernández-Veledo S, Grau-Bové C, Notararigo S, Huber-Ruano I. The role of microbial succinate in the pathophysiology of inflammatory bowel disease: mechanisms and therapeutic potential. Curr Opin Microbiol 2025; 85:102599. [PMID: 40132355 DOI: 10.1016/j.mib.2025.102599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/28/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-mediated condition linked to gut microbiota dysbiosis and altered production of bacterial metabolites, including succinate, which is also a key intermediate in human mitochondrial energy metabolism in human cells. Succinate levels in the gut are influenced by microbial community dynamics and cross-feeding interactions, highlighting its dual metabolic and ecological importance. Extracellular succinate acts as a key signaling metabolite linking microbial metabolism to host physiology, with transient rises supporting metabolic regulation but chronic elevations contributing to metabolic disorders and disease progression. Succinate signals through its cognate receptor SUCNR1, which mediates adaptive metabolic responses under normal conditions but drives inflammation and fibrosis when dysregulated. IBD patients display a dysbiotic gut microbiota characterized by an increased prevalence of succinate-producing bacteria, contributing to elevated succinate levels in the gut and circulation. This imbalance drives inflammation, worsens IBD severity, and contributes to complications like Clostridioides difficile infection and fibrosis. Emerging evidence highlights the potential of intestinal and systemic succinate levels as indicators of microbial dysbiosis, with a bidirectional relationship between microbial composition and succinate metabolism. Understanding the factors influencing succinate levels and their interaction with dysbiosis shows promise in the development of therapeutic strategies to restore microbial balance. Approaches such as dietary fiber enrichment, prebiotics, and probiotics to enhance succinate-consuming bacteria, combined with targeted modulation of succinate pathways (e.g. SDH inhibitors, SUCNR1 antagonists), hold promise for mitigating inflammation and improving gut health in IBD.
Collapse
Affiliation(s)
- Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Department de Ciències Mèdiques Bàsiques, University Rovira i Virgili, Tarragona, Spain.
| | - Carme Grau-Bové
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; SucciPro, S.L, Barcelona, Spain
| | - Sara Notararigo
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; SucciPro, S.L, Barcelona, Spain
| | - Isabel Huber-Ruano
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; SucciPro, S.L, Barcelona, Spain.
| |
Collapse
|
44
|
Tian M, Hao F, Jin X, Wang X, Chang T, He S, Wang H, Jiang Y, Wang Y, Liu J, Feng Y, Li D, Yin Z, Ba X, Wei M. KLHL25-ACLY module functions as a switch in the fate determination of the differentiation of iTreg/Th17. Commun Biol 2025; 8:471. [PMID: 40119138 PMCID: PMC11928475 DOI: 10.1038/s42003-025-07917-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
The differentiation of Th17 and iTreg is tightly associated with fatty acid metabolism. TGFβ1-induced iTreg differentiation from Th0 relies on fatty acid oxidation (FAO), whereas IL-6 with TGFβ1 shifts metabolism to Th17-preferred fatty acid synthesis (FAS). However, how IL-6 reprograms fatty acid metabolism remains unclear. Here, we unveiled that TGFβ1-activated JNK is recruited to the Klhl25 promoter by NF-YA. JNK then phosphorylates histone H3 at Ser10 to activate Klhl25 transcription, leading to the ubiquitination-dependent degradation of ATP-citrate lyase (ACLY) and the switch from FAS to FAO, which supports iTreg generation. Whereas, upon IL-6 signaling, NF-YA is phosphorylated by ERK, losing its DNA binding ability, which shuts off TGFβ1-JNK-mediated Klhl25 transcription and ACLY ubiquitination, thereby increasing FAS and supporting Th17 differentiation. This study demonstrated that KLHL25-ACLY module functions as a switch in response to TGFβ1 and IL-6 signals, playing a decisive role in the fate determination of iTreg/Th17 differentiation.
Collapse
Affiliation(s)
- Miaomiao Tian
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Fengqi Hao
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
- School of Physical Education, Northeast Normal University, Changchun, Jilin, China
| | - Xin Jin
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Xinyu Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Tianyi Chang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Shuang He
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Huiyue Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Ying Jiang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Yang Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Jia Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Yunpeng Feng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Dan Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhinan Yin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Zhuhai, China
| | - Xueqing Ba
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China.
| | - Min Wei
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China.
| |
Collapse
|
45
|
Oami T, Shimazui T, Yumoto T, Otani S, Hayashi Y, Coopersmith CM. Gut integrity in intensive care: alterations in host permeability and the microbiome as potential therapeutic targets. J Intensive Care 2025; 13:16. [PMID: 40098052 PMCID: PMC11916345 DOI: 10.1186/s40560-025-00786-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND The gut has long been hypothesized to be the "motor" of critical illness, propagating inflammation and playing a key role in multiple organ dysfunction. However, the exact mechanisms through which impaired gut integrity potentially contribute to worsened clinical outcome remain to be elucidated. Critical elements of gut dysregulation including intestinal hyperpermeability and a perturbed microbiome are now recognized as potential therapeutic targets in critical care. MAIN BODY The gut is a finely tuned ecosystem comprising ~ 40 trillion microorganisms, a single cell layer intestinal epithelia that separates the host from the microbiome and its products, and the mucosal immune system that actively communicates in a bidirectional manner. Under basal conditions, these elements cooperate to maintain a finely balanced homeostasis benefitting both the host and its internal microbial community. Tight junctions between adjacent epithelial cells selectively transport essential molecules while preventing translocation of pathogens. However, critical illness disrupts gut barrier function leading to increased gut permeability, epithelial apoptosis, and immune activation. This disruption is further exacerbated by a shift in the microbiome toward a "pathobiome" dominated by pathogenic microbes with increased expression of virulence factors, which intensifies systemic inflammation and accelerates organ dysfunction. Research has highlighted several potential therapeutic targets to restore gut integrity in the host, including the regulation of epithelial cell function, modulation of tight junction proteins, and inhibition of epithelial apoptosis. Additionally, microbiome-targeted therapies, such as prebiotics, probiotics, fecal microbiota transplantation, and selective decontamination of the digestive tract have also been extensively investigated to promote restoration of gut homeostasis in critically ill patients. Future research is needed to validate the potential efficacy of these interventions in clinical settings and to determine if the gut can be targeted in an individualized fashion. CONCLUSION Increased gut permeability and a disrupted microbiome are common in critical illness, potentially driving dysregulated systemic inflammation and organ dysfunction. Therapeutic strategies to modulate gut permeability and restore the composition of microbiome hold promise as novel treatments for critically ill patients.
Collapse
Affiliation(s)
- Takehiko Oami
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takashi Shimazui
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tetsuya Yumoto
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency, Critical Care and Disaster Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shunsuke Otani
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yosuke Hayashi
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA.
| |
Collapse
|
46
|
Canesso MC, Castro TB, Nakandakari-Higa S, Lockhart A, Luehr J, Bortolatto J, Parsa R, Esterházy D, Lyu M, Liu TT, Murphy KM, Sonnenberg GF, Reis BS, Victora GD, Mucida D. Identification of antigen-presenting cell-T cell interactions driving immune responses to food. Science 2025; 387:eado5088. [PMID: 39700315 PMCID: PMC12017586 DOI: 10.1126/science.ado5088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 08/09/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
The intestinal immune system must concomitantly tolerate food and commensals and protect against pathogens. Antigen-presenting cells (APCs) orchestrate these immune responses by presenting luminal antigens to CD4+ T cells and inducing their differentiation into regulatory (peripheral regulatory T cell) or inflammatory [T helper (Th) cell] subsets. We used a proximity labeling method (LIPSTIC) to identify APCs that presented dietary antigens under tolerizing and inflammatory conditions and to understand cellular mechanisms by which tolerance to food is induced and can be disrupted by infection. Helminth infections disrupted tolerance induction proportionally to the reduction in the ratio between tolerogenic APCs-including migratory dendritic cells (cDC1s) and Rorγt+ APCs-and inflammatory APCs, which were primarily cDC2s. These inflammatory cDC2s expanded by helminth infection did not present dietary antigens, thus avoiding diet-specific Th2 responses.
Collapse
Affiliation(s)
- Maria C.C. Canesso
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, United States
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, United States
| | - Tiago B.R. Castro
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, United States
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, United States
| | | | - Ainsley Lockhart
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, United States
| | - Julia Luehr
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, United States
| | - Juliana Bortolatto
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, United States
| | - Roham Parsa
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, United States
| | - Daria Esterházy
- Department of Pathology, University of Chicago, Chicago, United States
| | - Mengze Lyu
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Department of Microbiology and Immunology, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, United States
| | - Tian-Tian Liu
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, United States
| | - Kenneth M. Murphy
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, United States
| | - Gregory F. Sonnenberg
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Department of Microbiology and Immunology, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, United States
| | - Bernardo S. Reis
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, United States
| | - Gabriel D. Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, United States
- Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, United States
- Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| |
Collapse
|
47
|
Loddo F, Laganà P, Rizzo CE, Calderone SM, Romeo B, Venuto R, Maisano D, Fedele F, Squeri R, Nicita A, Nirta A, Genovese G, Bartucciotto L, Genovese C. Intestinal Microbiota and Vaccinations: A Systematic Review of the Literature. Vaccines (Basel) 2025; 13:306. [PMID: 40266208 PMCID: PMC11946530 DOI: 10.3390/vaccines13030306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 04/24/2025] Open
Abstract
Background: Vaccination constitutes a low-cost, safe, and efficient public health measure that can help prevent the spread of infectious diseases and benefit the community. The fact that vaccination effectiveness varies among populations, and that the causes of this are still unclear, indicates that several factors are involved and should be thoroughly examined. The "intestinal microbiota" is the most crucial of these elements. Numerous clinical studies demonstrate the intestinal microbiota's significance in determining the alleged "immunogenicity" and efficacy of vaccines. This systematic review aimed to review all relevant scientific literature and highlight the role of intestinal microbiota in COVID-19, Salmonella typhi, Vibrio cholerae, and rotavirus vaccinations. Materials and Methods: The MESH terms "vaccines" and "microbiota" were used to search the major scientific databases PubMed, SciVerse Scopus, Web of Knowledge, and the Cochrane Central Register of Controlled Clinical Trials. Results: Between February 2024 and October 2024, the analysis was conducted using electronic databases, yielding a total of 235 references. Finally, 24 RCTs were chosen after meeting all inclusion criteria: eight studies of COVID-19, two studies of Salmonella typhi, three studies of Vibrio cholerae, and eleven studies of rotavirus. Only six of these demonstrated good study quality with a Jadad score of three or four. Conclusions: According to the review's results, the intestinal microbiota surely plays a role in vaccinations' enhanced immunogenicity, especially in younger people. As it is still unclear what mechanisms underlie this effect, more research is needed to better understand the role of the intestinal microbiota.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Giovanni Genovese
- Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy; (F.L.); (P.L.); (C.E.R.); (S.M.C.); (B.R.); (R.V.); (D.M.); (F.F.); (R.S.); (A.N.); (A.N.); (L.B.)
| | | | - Cristina Genovese
- Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy; (F.L.); (P.L.); (C.E.R.); (S.M.C.); (B.R.); (R.V.); (D.M.); (F.F.); (R.S.); (A.N.); (A.N.); (L.B.)
| |
Collapse
|
48
|
Zeamer AL, Lai Y, Sanborn V, Loew E, Tracy M, Jo C, Ward DV, Bhattarai SK, Drake J, McCormick BA, Bucci V, Haran JP. Microbiome functional gene pathways predict cognitive performance in older adults with Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641911. [PMID: 40161798 PMCID: PMC11952313 DOI: 10.1101/2025.03.06.641911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Disturbances in the gut microbiome is increasing correlated with neurodegenerative disorders, including Alzheimer's Disease. The microbiome may in fact influence disease pathology in AD by triggering or potentiating systemic and neuroinflammation, thereby driving disease pathology along the "microbiota-gut-brain-axis". Currently, drivers of cognitive decline and symptomatic progression in AD remain unknown and understudied. Changes in gut microbiome composition may offer clues to potential systemic physiologic and neuropathologic changes that contribute to cognitive decline. Here, we recruited a cohort of 260 older adults (age 60+) living in the community and followed them over time, tracking objective measures of cognition, clinical information, and gut microbiomes. Subjects were classified as healthy controls or as having mild cognitive impairment based on cognitive performance. Those with a diagnosis of Alzheimer's Diseases with confirmed using serum biomarkers. Using metagenomic sequencing, we found that relative species abundances correlated well with cognition status (MCI or AD). Furthermore, gene pathways analyses suggest certain microbial metabolic pathways to either be correlated with cognitive decline or maintaining cognitive function. Specifically, genes involved in the urea cycle or production of methionine and cysteine predicted worse cognitive performance. Our study suggests that gut microbiome composition may predict AD cognitive performance.
Collapse
Affiliation(s)
- Abigail L. Zeamer
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yushuan Lai
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | - Ethan Loew
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Matthew Tracy
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Cynthia Jo
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Doyle V. Ward
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shakti K. Bhattarai
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | - Beth A. McCormick
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Vanni Bucci
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - John P. Haran
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
49
|
Engelhart MJ, Brock OD, Till JM, Glowacki RWP, Cantwell JW, Clarke DJ, Wesener DA, Ahern PP. BT1549 coordinates the in vitro IL-10 inducing activity of Bacteroides thetaiotaomicron. Microbiol Spectr 2025; 13:e0166924. [PMID: 39868786 PMCID: PMC11878027 DOI: 10.1128/spectrum.01669-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/27/2024] [Indexed: 01/28/2025] Open
Abstract
The intestine is home to a complex immune system that is engaged in mutualistic interactions with the microbiome that maintain intestinal homeostasis. A variety of immune-derived anti-inflammatory mediators have been uncovered and shown to be critical for maintaining these beneficial immune-microbiome relationships. Notably, the gut microbiome actively invokes the induction of anti-inflammatory pathways that limit the development of microbiome-targeted inflammatory immune responses. Despite the importance of this microbiome-driven immunomodulation, detailed knowledge of the microbial factors that promote these responses remains limited. We have previously established that the gut symbiont Bacteroides thetaiotaomicron stimulates the production of the anti-inflammatory cytokine IL-10 via soluble factors in a Toll-like receptor 2 (TLR2)-MyD88-dependent manner. Here, using TLR2 activity reporter cell lines, we show that the capacity of B. thetaiotaomicron to stimulate TLR2 activity was not critically dependent on either of the canonical heterodimeric forms of TLR2, TLR2/TLR1, or TLR2/TLR6, that typically mediate its function. Furthermore, biochemical manipulation of B. thetaiotaomicron-conditioned media suggests that IL-10 induction is mediated by a protease-resistant or non-proteogenic factor. We next uncovered that deletion of gene BT1549, a predicted secreted lipoprotein, significantly impaired the capacity of B. thetaiotaomicron to induce IL-10, while complementation in trans restored IL-10 induction, suggesting a role for BT1549 in the immunomodulatory function of B. thetaiotaomicron. Collectively, these data provide molecular insight into the pathways through which B. thetaiotaomicron operates to promote intestinal immune tolerance and symbiosis. IMPORTANCE Intestinal homeostasis requires the establishment of peaceful interactions between the gut microbiome and the intestinal immune system. Members of the gut microbiome, like the symbiont Bacteroides thetaiotaomicron, actively induce anti-inflammatory immune responses to maintain mutualistic relationships with the host. Despite the importance of such interactions, the specific microbial factors responsible remain largely unknown. Here, we show that B. thetaiotaomicron, which stimulates Toll-like receptor 2 (TLR2) to drive IL-10 production, can stimulate TLR2 independently of TLR1 or TLR6, the two known TLR that can form heterodimers with TLR2 to mediate TLR2-dependent responses. Furthermore, we show that IL-10 induction is likely mediated by a protease-resistant or non-proteogenic factor, and that this requires gene BT1549, a predicted secreted lipoprotein and peptidase. Collectively, our work provides insight into the molecular dialog through which B. thetaiotaomicron coordinates anti-inflammatory immune responses. This knowledge may facilitate future strategies to promote such responses for therapeutic purposes.
Collapse
Affiliation(s)
- Morgan J. Engelhart
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Orion D. Brock
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jessica M. Till
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Robert W. P. Glowacki
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jason W. Cantwell
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - David J. Clarke
- School of Microbiology and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Darryl A. Wesener
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Philip P. Ahern
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
50
|
Lu X, Xv Y, Hu W, Sun B, Hu H. Targeting CD4+ T cells through gut microbiota: therapeutic potential of traditional Chinese medicine in inflammatory bowel disease. Front Cell Infect Microbiol 2025; 15:1557331. [PMID: 40099014 PMCID: PMC11911530 DOI: 10.3389/fcimb.2025.1557331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
Inflammatory Bowel Disease (IBD) is an autoimmune disease characterized by chronic relapsing inflammation of the intestinal tract. Gut microbiota (GM) and CD4+T cells are important in the development of IBD. A lot of studies have shown that GM and their metabolites like short-chain fatty acids, bile acids and tryptophan can be involved in the differentiation of CD4+T cells through various mechanisms, which in turn regulate the immune homeostasis of the IBD patients. Therefore, regulating CD4+T cells through GM may be a potential therapeutic direction for the treatment of IBD. Many studies have shown that Traditional Chinese Medicine (TCM) formulas and some herbal extracts can affect CD4+T cell differentiation by regulating GM and its metabolites. In this review, we mainly focus on the role of GM and their metabolites in regulating the differentiation of CD4+T cells and their correlation with IBD. We also summarize the current research progress on the regulation of this process by TCM.
Collapse
Affiliation(s)
- Xingyao Lu
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yichuan Xv
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiye Hu
- Department of Liver Disease, Shanghai Yueyang Integrated Traditional Chinese Medicine and Western Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Boyun Sun
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongyi Hu
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|